1
|
Wu L, Sun W, Huang L, Sun L, Dou J, Lu G. Calcium Imaging in Vivo: How to Correctly Select and Apply Fiber Optic Photometric Indicators. Organogenesis 2025; 21:2489667. [PMID: 40186873 PMCID: PMC11980459 DOI: 10.1080/15476278.2025.2489667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/11/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025] Open
Abstract
Fiber-photometric is a novel optogenetic method for recording neural activity in vivo, which allows the use of calcium indicators to observe and study the relationship between neural activity and behavior in free-ranging animals. Calcium indicators also convert changes in calcium concentration in cells or tissues into recordable fluorescent signals, which can then be observed using the system of fiber-photometric. To date, there is a paucity of relevant literature on the proper selection and application of fiber-photometric indicators. Therefore, this paper will detail how to correctly select and apply fiber-photometer indicators in four sections: the basic principle of optical fiber photometry, the selection of calcium fluorescent probes and viral vector systems, and the measurement of specific expression of fluorescent proteins in specific tissues. Therefore, the correct use of suitable fiber optic recording indicators will greatly assist researchers in exploring the link between neuronal activity and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lanxia Wu
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, China
| | - Wenxuan Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Linjie Huang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, China
| | - Jinhua Dou
- Mental Health Education Center, Shandong Second Medical University, Weifang, Shandong, China
| | - Guohua Lu
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
2
|
Xu B, Yang L, Jiang R, Tao G, Zhi S, Sun L, Wu Y, Shi Y. Discovery of a novel quinoline RIP1 inhibitor and its treatment of acute liver injury in mice. Bioorg Chem 2025; 159:108365. [PMID: 40088687 DOI: 10.1016/j.bioorg.2025.108365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Necroptosis is closely associated with the development of inflammatory diseases, including acute liver injury. However, the precise role of necroptosis-related signature proteins in acute liver injury remains incompletely understood. Previously, our group investigated Compound o1, a RIP1 inhibitor, but its antinecroptosis activity and RIP1 binding affinity were suboptimal. In this study, we sought to address these two critical scientific challenges. Through a scaffold-hopping strategy, we identified a series of novel quinoline-like RIP1 inhibitors, among which N-1 exhibited the most potent antinecroptosis activity and the strongest RIP1 binding affinity. N-1 effectively inhibited necrosome formation by blocking phosphorylation in the RIP1/RIP3/MLKL signaling pathway. In a TNF-induced hypothermia mouse model of systemic inflammatory response syndrome (SIRS), N-1 significantly improved the survival rate of mice in a dose-dependent manner. Our study further revealed that RIP1, RIP3, and MLKL are expressed in normal liver tissues, whereas their phosphorylated forms (pRIP1, pRIP3, and pMLKL) are absent. In contrast, liver tissues from mice with CCl4-induced acute liver injury exhibited high expression levels of pRIP1, pRIP3, and pMLKL, indicating that necroptosis is associated with liver injury. N-1 significantly inhibited the phosphorylation of RIP1, RIP3, and MLKL, while restoring key liver damage markers such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST). These findings suggest that targeting necroptosis may represent a promising therapeutic strategy for the treatment of acute liver injury.
Collapse
Affiliation(s)
- Bin Xu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Linghui Yang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Ruiqi Jiang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Guojing Tao
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Shumeng Zhi
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Lei Sun
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Yanran Wu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China.
| | - Ying Shi
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China.
| |
Collapse
|
3
|
Ameri A, Gandomkar H, Ahmed HH, Kareem RA, Sameer HN, Yaseen A, Athab ZH, Adil M, Ghasemzadeh I. A review of the progress and challenges of developing dendritic-based vaccines against hepatitis B virus (HBV). Pathol Res Pract 2025; 271:156025. [PMID: 40382895 DOI: 10.1016/j.prp.2025.156025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/13/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Hepatitis B virus (HBV) infections that last a long time are a significant public health problem worldwide. About 254 million people around the world are chronically sick with HBV. Each year, 1.2 million new cases occur, and in 2022, 1.1 million people will die from the disease. So, it has been essential to work on finding ways to treat and avoid HBV. The process of therapeutic vaccination involves giving people a non-infectious form of a virus to start or improve immune reactions specific to HBV. This helps keep HBV infections under control. Dendritic cells (DCs) play a significant part in beginning the adaptive immune response, which could decide how well an HBV infection is treated. DC-based treatment has been looked into for people with chronic HBV (CHB) infection and has shown some sound effects. Vaccines for CHB that use DCs boost antiviral immunity by improving T cells and breaking the immune system's resistance against HBV. In these vaccines, DCs are loaded with HBV antigens (like HBsAg, HBcAg, or peptides) outside of the body and then put back into the patient to make the immune system work better. In conclusion, this DC treatment is a biological therapy method with a good chance of being used. This study examined the different DC-based medicines that can treat and prevent HBV. Finally, we've talked about clinical studies, the current problems, how to fix them, and the future of this vaccine for treating and preventing HBV.
Collapse
Affiliation(s)
- Ali Ameri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hossein Gandomkar
- Department of Surgical Oncology, Tehran University of Medical Medicine, Tehran, Iran
| | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | | - Iman Ghasemzadeh
- Research Center Of Tropical and Infectious Diseases, Kerman University Of Medical Sciences, Kerman, Iran.
| |
Collapse
|
4
|
Wang JW, Zhu HX, Zhang F, Wang H, Fan YC, Han LY, Wang K. Reduced murine double minute-2 methylation from peripheral blood mononuclear cells correlates with enhanced oxidative stress in hepatitis b virus-related hepatocellular carcinoma. Front Microbiol 2025; 16:1590492. [PMID: 40432974 PMCID: PMC12106469 DOI: 10.3389/fmicb.2025.1590492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Background Hepatitis B virus-related hepatocarcinogenesis (HBV-related HCC) involves a variety of causes including oncogene hypomethylation, oxidative stress and HBV itself. Oxidative stress induces an alternation in the DNA methylation status. We aimed to study the relationship between oxidative stress and murine double minute-2 (MDM2) methylation status in HBV-related HCC patients and healthy controls (HCs). Methods A total of 135 patients with HBV-related HCC and 26 healthy controls (HCs) were recruited. The MDM2 methylation status was detected by methylation-specific PCR. The expression of MDM2 mRNA was assessed using quantitative real-time PCR. The plasma malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), nuclear factor erythroid 2-related factor 2 (NRF2), heme Oxygenase-1 (HO-1), and glutathione peroxidase 4 (GPX4) were measured by enzyme-linked immunosorbent assay (ELISA). Thirty-six patients with HBV-related HCC and 11 HCs were selected and the serum metabolism was analyzed by ultra-high-performance liquid chromatography-mass spectrometry (UHPLC-MS). Results Compared with HCs, the MDM2 promoter methylation frequency was significantly decreased in HBV-related HCC. The MDA levels were increased, whereas the GSH, SOD, NRF2, HO-1, and GPX4 levels were decreased in the HBV-related HCC patients relative to HCs. There were 216 differential metabolites between HBV-related HCC and HCs in plasma, which belongs to amino acids, bile acids, fatty acids, phospholipids, and other compounds. The cysteine and methionine metabolism were the most significant metabolic pathways associated with differential metabolites between MDM2 methylated group and MDM2 unmethylated group in HBV-related HCC. Conclusion Our results suggested that oxidative stress may cause MDM2 hypomethylation, in which cysteine and methionine pathway might play an important role in.
Collapse
Affiliation(s)
- Jing-Wen Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Han-Xu Zhu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Feng Zhang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - He Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Li-Yan Han
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| |
Collapse
|
5
|
Ling S, Yu J, Zhan Q, Gao M, Liu P, Wu Y, Zhang L, Shan Q, Liu H, Wang J, Cai S, Zhou W, Que Q, Wang S, Hong J, Xiang J, Xu S, Liu J, Peng X, Wang N, Wang W, Xie H, Cai J, Wang L, Zheng S, Xu X. Multi-omic analysis reveals a CAF-stemness-governed classification in HCC liver transplant recipients beyond the Milan criteria. Nat Commun 2025; 16:4392. [PMID: 40355422 PMCID: PMC12069600 DOI: 10.1038/s41467-025-59745-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
In patients with hepatocellular carcinoma (HCC) meeting the Milan criteria, liver transplantation (LT) is an effective therapy. This study aims to define the survival-related molecular biological features helping precisely identifying the patients with HCC beyond the Milan criteria who have acceptable outcomes. In the derivation cohort, integrated analyses of tumor tissues are conducted using RNA sequencing (RNA-seq), proteomic landscape, and transposase-accessible chromatin sequencing (ATAC-seq). Based on transcriptomics, three subgroups that significantly differ in overall survival were identified in the derivation cohort, and these findings are validated in an independent cohort. In-depth bioinformatics analysis using RNA-seq and proteomics reveals that the promotion of cancer stemness by cancer-associated fibroblasts (CAFs) can be responsible for the negative biological characteristics observed in high-risk HCC patients. The ATAC-seq identifies key factors regulating transcription, which may bridge CAF infiltration and stemness. Finally, we demonstrate that the CAF-derived CXCL12 sustains the stemness of HCC cells by promoting XRCC5 through CXCR4.
Collapse
Affiliation(s)
- Sunbin Ling
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.
| | - Jiongjie Yu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Qifan Zhan
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Mingwei Gao
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Peng Liu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yongfeng Wu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Lincheng Zhang
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Qiaonan Shan
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huan Liu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiapei Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Shuqi Cai
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Wei Zhou
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Qingyang Que
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Shuo Wang
- Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Jiachen Hong
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Jianan Xiang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Shengjun Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Jimin Liu
- Department of Pathology and Laboratory Medicine, Mt Sinai Hospital, Toronto, ON, Canada
| | - Xiaojun Peng
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Nan Wang
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Weixin Wang
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Haiyang Xie
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Jinzhen Cai
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Liming Wang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China.
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China.
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.
| |
Collapse
|
6
|
Lavoro A, Ricci D, Gattuso G, Longo F, Spoto G, Vitale ACV, Giuliana MC, Falzone L, Libra M, Candido S. Recent advances on gene-related DNA methylation in cancer diagnosis, prognosis, and treatment: a clinical perspective. Clin Epigenetics 2025; 17:76. [PMID: 40325471 PMCID: PMC12054201 DOI: 10.1186/s13148-025-01884-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/13/2025] [Indexed: 05/07/2025] Open
Abstract
Recent advances in screening programs and the development of innovative therapeutic strategies have significantly improved the clinical outcomes of cancer patients. However, many patients still experience treatment failure, primarily due to inherent or acquired drug resistance mechanisms. This challenge underscores the urgent need for novel therapeutic targets for the effective treatment of malignancies, as well as cancer-specific biomarkers to enhance early diagnosis and guide interventions. Epigenetic mechanisms, including DNA methylation, have recently garnered growing interest as key regulators of gene expression under both physiological and pathological conditions. Although epigenetic dysregulations are reliable tumor hallmarks, DNA methylation is still not routinely integrated into clinical practice, highlighting the need for further research to translate preclinical findings from the bench to the bedside. On these bases, the present review aims to illustrate the state of the art regarding the role of DNA methylation in cancer, describing the technologies currently available for DNA methylation profiling. Furthermore, the latest evidence on the application of DNA methylation hotspots in cancer diagnosis and prognosis, as well as the impact of epidrugs in cancer care, is discussed to provide a comprehensive overview of the potential clinical relevance of DNA methylation in advancing personalized medicine.
Collapse
Affiliation(s)
- Alessandro Lavoro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Daria Ricci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Federica Longo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Graziana Spoto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | | | - Maria Chiara Giuliana
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy.
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123, Catania, Italy
| |
Collapse
|
7
|
Wang G, Cao Y, Hu T, Cai Z, Chen C, Geng Q, Luo X, Liu Y, Wang W, Jin J, Sheng W. A Mutual Interaction Between GSTP1 and p53 Improves the Drug Resistance and Malignant Biology of Pancreatic Cancer. Cancer Sci 2025; 116:1268-1281. [PMID: 39953720 PMCID: PMC12044643 DOI: 10.1111/cas.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
Glutathione S-transferase P1 (GSTP1), a classic tumor biomarker, plays a controversial role in cancer progression. However, its specific role in pancreatic cancer (PC) has rarely been investigated. In the present study, we investigated the function and relationship between GSTP1 and mutant/wild-type p53 (mtp53/wtp53) in PC in vitro and in vivo. Compared with paired adjacent normal pancreas tissue, GSTP1 was downregulated in PC tissue, which was closely correlated with lymph node metastasis, Union for International Cancer Control (UICC) stage, and a better outcome of PC patients, processes dependent on wtp53 rather than mtp53. Moreover, a mutual regulation between GSTP1 and p53 was found in wtp53 PC cells. GSTP1 overexpression inhibited cell proliferation and chemotherapy resistance in vitro via wtp53/p21 and Bax/Bcl2 signaling, which was significantly reversed by wtp53 silencing, and vice versa. Similarly, the coordination of GSTP1 and p53 regulated the invasion and migration of PC cells, which was accompanied by changes in epithelial-mesenchymal transition (EMT) signaling (E-cad, ZO-1 and MMP9). Moreover, GSTP1 overexpression inhibited tumor growth and liver metastasis in vivo, as did high wtp53 and low ki67 expression. Interestingly, GSTP1 did not coimmunoprecipitate with either mtp53 or wtp53 in vitro. However, the wtp53 protein, as a transcription factor, could bind to the GSTP1 DNA promoter to transactivate GSTP1 mRNA expression as demonstrated via a Chip assay. Additionally, GSTP1 promoted the translocation of wtp53 into the nucleus but not mtp53. These results suggest that the positive feedback regulation of GSTP1 and wtp53 plays a significant role in cell proliferation, drug resistance, cell invasion and metastasis in PC.
Collapse
Affiliation(s)
- Guosen Wang
- Department of General SurgeryThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Yi Cao
- Department of General SurgeryThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Tengcheng Hu
- Department of General SurgeryThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Zhengqing Cai
- Department of General SurgeryThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - ChuanPing Chen
- Department of PharmacyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Qilong Geng
- Department of Clinical MedicineThe First Clinical College, Anhui Medical UniversityHefeiChina
| | - Xinyu Luo
- Department of Clinical MedicineThe First Clinical College, Anhui Medical UniversityHefeiChina
| | - Yang Liu
- Department of Clinical MedicineThe First Clinical College, Anhui Medical UniversityHefeiChina
| | - Weijie Wang
- Department of Clinical MedicineThe First Clinical College, Anhui Medical UniversityHefeiChina
| | - Jiabin Jin
- Department of General SurgeryPancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of General SurgeryRuijin‐Hainan Hospital, Shanghai Jiao Tong University School of MedicineQionghaiChina
| | - Weiwei Sheng
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
8
|
Yang K, Lee J, Han JW, Yang H, Chae SY, Chung BS, Ryu T. A comprehensive analysis of the impact of smoking on adverse clinical outcomes of steatotic liver diseases. Therap Adv Gastroenterol 2025; 18:17562848251331315. [PMID: 40292092 PMCID: PMC12033444 DOI: 10.1177/17562848251331315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is an increasingly prevalent liver disorder. Objectives This study investigated the effect of smoking status on various clinical outcomes in MASLD and metabolic dysfunction and alcohol-associated liver disease (MetALD). Design This study is a retrospective cohort analysis utilizing data from the UK Biobank (Application ID: 117214). Participants were categorized as current, previous, or never smokers, and outcomes were analyzed using inverse probability of treatment weighting to adjust for confounders. Methods The primary outcomes were all-cause mortality and liver-related mortality. Secondary outcomes included incidence of liver cirrhosis, hepatic decompensation, cardio-cerebrovascular diseases (CVD), and hepatocellular carcinoma (HCC). Multivariate Cox proportional hazards models were employed to evaluate associations. Results Previous and never smokers had significantly lower hazard ratios (HRs) for mortality compared to current smokers in all cohorts (HR: 0.33, 95% confidence interval (CI): 0.31-0.35, p < 0.001 for never smokers in No SLD cohort, HR: 0.43, 95% CI: 0.41-0.44, p < 0.001 for never smokers in MASLD cohort, and HR: 0.41, 95% CI: 0.38-0.45, p < 0.001 for never smokers in MetALD cohort). Previous and never smokers showed significantly lower incidences of liver cirrhosis compared to current smokers across all cohorts, except for MetALD. Previous and never smokers showed lower incidences of CVD compared to current smokers. In the MASLD cohort, never smokers had the lowest incidence of hepatic decompensation and HCC. In the MetALD cohort, no significant differences were observed in the risk of hepatic decompensation and HCC between different smoking statuses. Conclusion Smoking is related to worse survival outcomes and higher incidences of liver cirrhosis and CVD in MASLD and MetALD cohorts. Therefore, smoking cessation and prevention are crucial strategies for reducing the burden of liver disease and improving patient prognosis.
Collapse
Affiliation(s)
- Keungmo Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jaejun Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Won Han
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Yun Chae
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Beom Sun Chung
- Department of Anatomy, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Tom Ryu
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, 59 Daesagwan-ro, Yongsan-gu, Seoul 04401, Republic of Korea
| |
Collapse
|
9
|
Wang R, Wang S, Mi Y, Huang T, Wang J, Ni J, Wang J, Yin J, Li M, Ran X, Fan S, Sun Q, Tan SY, Phillip Koeffler H, Ding L, Chen YQ, Feng N. Elevated serum levels of GPX4, NDUFS4, PRDX5, and TXNRD2 as predictive biomarkers for castration resistance in prostate cancer patients: an exploratory study. Br J Cancer 2025; 132:543-557. [PMID: 39900986 PMCID: PMC11920399 DOI: 10.1038/s41416-025-02947-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 12/24/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a heterogeneous disease affecting over 14% of the male population worldwide. Although patients often respond positively to initial treatments within the first 2-3 years, many eventually develop a more lethal form of the disease known as castration-resistant PCa (CRPC). At present, no biomarkers that predict the onset of CRPC are available. This study aims to provide insights into the diagnosis and prediction of CRPC emergence. METHODS Protein expression dynamics were analysed in drug (androgen receptor inhibitor)-tolerant persister (DTP) and drug withdrawal cells using proteomics to identify potential biomarkers. These biomarkers were subsequently validated using a mouse model, 180-paired carcinoma/benign tissues, and 482 serum samples. Five machine learning algorithms were employed to build clinical prediction models, wherein the SHapley Additive exPlanation (SHAP) framework was used to interpret the best-performing model. Moreover, three regression models were developed to determine the Time from initial PCa diagnosis to CRPC development (TPC) in patients. RESULTS We identified that the protein expression levels of GPX4, NDUFS4, PRDX5, and TXNRD2 were significantly upregulated in PCa patients, particularly in those with CRPC. Among the tested machine learning models, the random forest and extreme gradient boosting models performed best on tissue and serum cohorts, achieving AUCs of 0.958 and 0.988, respectively. In addition, a significant inverse correlation was observed between TPC and serum levels of these four biomarkers. This correlation was formulated in three regression models, which achieved the smallest mean absolute error of 1.903 on independent datasets for predicting CRPC emergence. CONCLUSION Our study provides new insights into the role of DTP cells in CRPC development. The quad protein panel identified in our study, along with the post hoc and intrinsically explainable prediction models, may serve as a convenient and real-time prognostic tool, addressing the current lack of clinical biomarkers for CRPC.
Collapse
Affiliation(s)
- Rong Wang
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shaopeng Wang
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China
| | - Yuanyuan Mi
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Tianyi Huang
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Jun Wang
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jiang Ni
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jian Wang
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology & School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Menglu Li
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China
- Department of Urology, Wuxi No.2 People's Hospital, Nanjing Medical University, Wuxi, China
| | - Xuebin Ran
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shuangyi Fan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Qiaoyang Sun
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Soo Yong Tan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, California, Los Angeles, CA, USA
| | - Lingwen Ding
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Yong Q Chen
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| | - Ninghan Feng
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
- Department of Urology, Wuxi No.2 People's Hospital, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
10
|
Tocci NX, Wehrle CJ, Sun K, Jiao C, Hong H, Gross A, Allkushi E, Uysal M, Linganna MW, Stackhouse K, Hashimoto K, Schlegel A, Walsh R, Miller C, Kwon DCH, Aucejo F. Circulating tumor DNA in management of primary liver malignancy: A review of the literature and future directions. J Surg Oncol 2025; 131:879-887. [PMID: 39155663 PMCID: PMC12120392 DOI: 10.1002/jso.27825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/14/2024] [Indexed: 08/20/2024]
Abstract
Primary liver malignancies are a serious and challenging global health concern. The most common primary tumors are hepatocellular carcinoma and cholangiocarcinoma. These diseases portend poor prognosis when presenting with progressive, extensive disease. There is a critical need for improved diagnosis, therapeutic intervention, and monitoring surveillance in liver-related malignancies. Liquid biopsy using ctDNA provides an opportunity for growth within these domains for liver-related malignancy. However, ctDNA is relatively understudied in this field compared with other solid tumor types, possibly due to the complex nature of the pathology. In this review, we aim to discuss ctDNA, the current literature, and future directions of this technology within primary liver malignancies.
Collapse
Affiliation(s)
- Noah X. Tocci
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Chase J. Wehrle
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Keyue Sun
- Lerner Research Institute, Inflammation & Immunity, Cleveland Clinic FoundationClevelandOhioUSA
| | - Chunbao Jiao
- Lerner Research Institute, Inflammation & Immunity, Cleveland Clinic FoundationClevelandOhioUSA
| | - Hanna Hong
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Abby Gross
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Erlind Allkushi
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Melis Uysal
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Maureen Whitsett Linganna
- Department of Gastroenterology, Hepatology, and NutritionDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Katheryn Stackhouse
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Koji Hashimoto
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Andrea Schlegel
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
- Lerner Research Institute, Inflammation & Immunity, Cleveland Clinic FoundationClevelandOhioUSA
| | - R. Matthew Walsh
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Charles Miller
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - David C. H. Kwon
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Federico Aucejo
- Department of Hepato‐pancreato‐biliary & Liver Transplant SurgeryDigestive Diseases and Surgery Institute, Cleveland Clinic FoundationClevelandOhioUSA
| |
Collapse
|
11
|
Chen N, Luo P, Tang Y, Liu P, Wang J, Fan Y, Han L, Wang K. Accelerators of chronic hepatitis B fibrosis cirrhosis CCND1 gene expression and promoter hypomethylation. Sci Rep 2025; 15:10630. [PMID: 40148411 PMCID: PMC11950333 DOI: 10.1038/s41598-025-93778-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
This study investigates the relationship between Cyclin D1 (CCND1) gene and promoter methylation and liver fibrosis (LF)/liver cirrhosis (LC)induced by chronic hepatitis B (CHB). Peripheral blood mononuclear cells (PBMCs) are collected from patients diagnosed with chronic hepatitis B (CHB) and hepatitis B-related LF/LC, as well as from healthy individuals. The mRNA levels and promoter methylation of the CCND1 gene are measured. Single-cell analysis is performed to determine the cell types primarily expressing the CCND1 gene in LF/LC. The GSE84044 dataset is utilized to validate the experimental results. Single-gene GSEA and immune infiltration analyses are conducted to identify significant pathways and immune characteristics associated with the CCND1 gene. The mRNA level of CCND1 in PBMCs from patients with hepatitis B-related LF/LC is elevated compared to those with chronic hepatitis B (CHB) and healthy individuals, while the promoter methylation level of CCND1 is reduced. Single-cell analysis indicates high expression of CCND1 in M2 macrophages (M2) and T cells. The GSE84044 dataset confirms higher CCND1 mRNA levels in liver tissues from patients with CHB-related LF/LC compared to CHB patients. Single-gene GSEA analysis associates CCND1 expression with natural killer cell-mediated cytotoxicity, T cell receptor signaling, and B cell receptor signaling pathways. Increased expression of CCND1 enhances immune infiltration during the fibrosis/cirrhosis process of CHB. The CCND1 expression and promoter methylation may be involved in the process of LF/LC in CHB and may be related to the immune response in the course of the disease.
Collapse
Affiliation(s)
- Nan Chen
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Pengyu Luo
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Yuna Tang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Pei Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Jing Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Yuchen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
- Institute of Hepatology, Shandong University, Jinan, 250012, People's Republic of China
| | - Liyan Han
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China.
- Institute of Hepatology, Shandong University, Jinan, 250012, People's Republic of China.
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China.
- Institute of Hepatology, Shandong University, Jinan, 250012, People's Republic of China.
| |
Collapse
|
12
|
Papatheodoridi A, Lekakis V, Chatzigeorgiou A, Papatheodoridis G. The Current Role of Circulating Cell-Free DNA in the Management of Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:1042. [PMID: 40149374 PMCID: PMC11940583 DOI: 10.3390/cancers17061042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Circulating cell-free DNA (cfDNA) has emerged as a compelling candidate of liquid biopsy markers for the diagnosis and prognosis of several cancers. We systematically reviewed data on the role of cfDNA markers in the diagnosis, prognosis and treatment of hepatocellular carcinoma (HCC). Early studies suggested that levels of circulating cfDNA, mitochondrial DNA and cfDNA integrity are higher in patients with HCC than chronic liver diseases. In subsequent studies, methylation changes in circulating tumor DNA (ctDNA) as well as cfDNA fragmentation patterns and circulating nucleosomes were found to offer high sensitivity (>60%) and excellent specificity (>90%) for HCC diagnosis. The predictive role of cfDNA markers and ctDNA has been assessed in a few studies including untreated patients with HCC providing promising results for prediction of survival. However, port-hepatectomy detection of cfDNA/ctDNA markers or copy number variation indicators of cfDNA seem to reflect minimum residual disease and thus a high risk for HCC recurrence. The same markers can be useful for prediction after transarterial chemoembolization, radiofrequency ablation, radiotherapy and even systemic therapies. In conclusion, cfDNA markers can be useful in HCC surveillance, improving early diagnosis rates, as well as for monitoring treatment effectiveness and minimal residual disease post-treatment.
Collapse
Affiliation(s)
- Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, “Alexandra” General Hospital of Athens, 11528 Athens, Greece;
| | - Vasileios Lekakis
- First Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - George Papatheodoridis
- First Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece;
| |
Collapse
|
13
|
Li JH, Tang Y, Wang J, Wei XF, Wang N, Wang JW, Lyu H, Jiang XM, Liu HH, Wang K. A Clinical Predictive Model Based on SOCS3 Promoter Methylation to Predict the Prognosis of Acute-on-Chronic Hepatitis B Liver Failure. J Inflamm Res 2025; 18:3741-3756. [PMID: 40109653 PMCID: PMC11920633 DOI: 10.2147/jir.s506050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE The study aimed to quantitatively detect the suppressors of cytokine signaling (SOCS) 3 promoter methylation levels, investigate the relationship between SOCS3 methylation and gene expression, and construct a prognosis prediction model combined with clinical indicators for Acute-on-chronic Hepatitis B Liver Failure (ACHBLF). METHODS A total of 135 ACHBLF patients were enrolled and randomly divided into the training cohort and validation cohort. The SOCS3 mRNA and promoter methylation in peripheral blood mononuclear cells (PBMCs) of ACHBLF patients were quantitative measured. A clinical prediction model was established based on SOCS3 promoter methylation and clinical indicators. The prediction model was evaluated by the area under the receiver operating characteristic curve, the Hosmer-Lemeshow (H-L) goodness-of-fit test, and decision curve analysis. RESULTS In this study, compared with ACHBLF survivals, SOCS3 showed lower mRNA levels and higher methylation levels in ACHBLF non-survivals. The SOCS3 methylation rates were negatively correlated with SOCS3 mRNA levels. PT-INR, IL-6, and percentage of the methylation reference (PMR) value (SOCS3) were used to establish a clinical model for predicting ACHBLF patients' prognosis. The results of AUC, the Hosmer-Lemeshow (H-L) goodness-of-fit test and decision curve analysis (DCA) showed that the prediction model had good clinical applicability. The prediction model was visualized. CONCLUSION A prognosis prediction model for ACHBLF was developed based on PMR (SOCS3), PT-INR and IL-6, which may have a good potential clinical application value.
Collapse
Affiliation(s)
- Ji-Hui Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yuna Tang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Jing Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xue-Fei Wei
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Na Wang
- Department of Hepatology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, People’s Republic of China
| | - Jing-Wei Wang
- Department of Hepatology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, People’s Republic of China
| | - Hui Lyu
- Department of Severe Liver Disease, Shandong Public Health Clinical Center of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xue-Mei Jiang
- Department of Hepatology, Shandong Public Health Clinical Center of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Hui-Hui Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Institute of Hepatology, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Institute of Hepatology, Shandong University, Jinan, Shandong, People’s Republic of China
- Shenzhen Research Institute, Shandong University, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
14
|
Ji Y, Xiao C, Fan T, Deng Z, Wang D, Cai W, Li J, Liao T, Li C, He J. The epigenetic hallmarks of immune cells in cancer. Mol Cancer 2025; 24:66. [PMID: 40038722 PMCID: PMC11881328 DOI: 10.1186/s12943-025-02255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the dysregulation of epigenetic mechanisms in cancer has emerged as a promising therapeutic strategy. Although the significant rationale progress of epigenetic therapies in blocking cancer cells, how epigenetic regulation shapes tumor microenvironment (TME) and establishes antitumor immunity remains less understood. Recent study focus has been put on the epigenetic-mediated changes in the fate of immune cells, including the differentiation, expansion, recruitment, functionalization, and exhaustion of T cells, natural killer (NK) cells, tumor-associated macrophages (TAMs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), and B cells within the TME. Here, we review the latest molecular and clinical insights into how DNA modifications, histone modification, and epitranscriptome-related regulations shape immune cells of various cancers. We also discuss opportunities for leveraging epigenetic therapies to improve cancer immunotherapies. This review provides the epigenetic foundations of cancer immunity and proposes the future direction of combination therapies.
Collapse
Affiliation(s)
- Yu Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianle Liao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
15
|
Babaeenezhad E, Khosravi P, Moradi Sarabi M. Dietary polyunsaturated fatty acids affect PPARγ promoter methylation status and regulate the PPARγ/COX2 pathway in some colorectal cancer cell lines. GENES & NUTRITION 2025; 20:2. [PMID: 40038577 DOI: 10.1186/s12263-025-00764-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Promoter methylation silencing of peroxisome proliferator-activated receptor gamma (PPARγ) and dysregulation of the PPARγ/COX2 axis contribute to colorectal cancer (CRC) pathogenesis. This study investigated for the first time the effects of dietary polyunsaturated fatty acids (PUFAs) on promoter methylation of PPARγ and the PPARγ/COX2 axis in five CRC cell lines. METHODS Five CRC cell lines (SW742, HCT116, Caco2, LS180, and HT29/219) were treated with 100 µM of eicosapentaenoic acid (EPA) or docosahexaenoic acid (DHA) or linoleic acid (LA). The methylation patterns of the four regions within the PPARγ promoter were determined using methylation-specific PCR (MSP). Additionally, the mRNA expression levels of PPARγ and COX2 were examined using RT-qPCR. RESULTS Our results showed that M3 segment within the PPARγ promoter was hemimethylated in SW742 cells, whereas other cell lines remained unmethylated in this region. The M4 region was hemimethylated in all the CRC cell lines. Of all PUFAs, DHA demethylated the M3 region of the PPARγ promoter in SW742 cells and the M4 region in Caco2 cells. Functionally, these changes were accompanied by significant upregulation of PPARγ in SW742 (9.22-fold; p = 0.01) and Caco2 cells (8.87-fold; p = 0.04). Additionally, COX2 expression was significantly downregulated in all CRC cell lines after exposure to PUFAs (p < 0.05). CONCLUSIONS This study demonstrated that PUFAs, particularly DHA, altered PPARγ promoter methylation and expression, as well as modulated the PPARγ/COX2 axis in CRC cells in a cell type-dependent manner. DHA was more effective than the other PUFAs in regulating PPARγ promoter methylation. Our results highlight the potential clinical use of PUFAs in CRC treatment.
Collapse
Affiliation(s)
- Esmaeel Babaeenezhad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Peyman Khosravi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mostafa Moradi Sarabi
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
- Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
16
|
Padhan P, Simran, Kumar N, Verma S. Glutathione S-transferase: A keystone in Parkinson's disease pathogenesis and therapy. Mol Cell Neurosci 2025; 132:103981. [PMID: 39644945 DOI: 10.1016/j.mcn.2024.103981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder that predominantly affects motor function due to the loss of dopaminergic neurons in the substantia nigra. It presents significant challenges, impacting millions worldwide with symptoms such as tremors, rigidity, bradykinesia, and postural instability, leading to decreased quality of life and increased morbidity. The pathogenesis of Parkinson's disease is multifaceted, involving complex interactions between genetic susceptibility, environmental factors, and aging, with oxidative stress playing a central role in neuronal degeneration. Glutathione S-Transferase enzymes are critical in the cellular defense mechanism against oxidative stress, catalysing the conjugation of the antioxidant glutathione to various toxic compounds, thereby facilitating their detoxification. Recent research underscores the importance of Glutathione S-Transferase in the pathophysiology of Parkinson's disease, revealing that genetic polymorphisms in Glutathione S-Transferase genes influence the risk and progression of the disease. These genetic variations can affect the enzymatic activity of Glutathione S-Transferase, thereby modulating an individual's capacity to detoxify reactive oxygen species and xenobiotics, which are implicated in Parkinson's disease neuropathological processes. Moreover, biochemical studies have elucidated the role of Glutathione S-Transferase in not only maintaining cellular redox balance but also in modulating various cellular signalling pathways, highlighting its neuroprotective potential. From a therapeutic perspective, targeting Glutathione S-Transferase pathways offers promising avenues for the development of novel treatments aimed at enhancing neuroprotection and mitigating disease progression. This review explores the evident and hypothesized roles of Glutathione S-Transferase in Parkinson's disease, providing a comprehensive overview of its importance and potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- Pratyush Padhan
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Simran
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeraj Kumar
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
17
|
Zhang S, Dong H, Jin X, Sun J, Li Y. The multifaceted roles of macrophages in the transition from hepatitis to hepatocellular carcinoma: From mechanisms to therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167676. [PMID: 39828046 DOI: 10.1016/j.bbadis.2025.167676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Macrophages are central to the progression from hepatitis to hepatocellular carcinoma (HCC), with their remarkable plasticity and ability to adapt to the changing liver microenvironment. Chronic inflammation, fibrosis, and ultimately tumorigenesis are driven by macrophage activation, making them key regulators of liver disease progression. This review explores the diverse roles of macrophages in the transition from hepatitis to HCC. In the early stages of hepatitis, macrophages are essential for pathogen clearance and tissue repair. However, chronic activation leads to prolonged inflammation, which exacerbates liver damage and promotes fibrosis. As the disease progresses to liver fibrosis, macrophages interact with hepatic stellate cells, fostering a pro-tumorigenic microenvironment that supports HCC development. In hepatocarcinogenesis, macrophages contribute to tumor initiation, growth, metastasis, immune evasion, cancer stem cell maintenance, and angiogenesis. Their functional plasticity enables them to adapt to the tumor microenvironment, thereby promoting tumor progression and resistance to therapy. Targeting macrophages represents a promising strategy for preventing and treating HCC. Therapeutic approaches, including reprogramming macrophage phenotypes to enhance anti-tumor immunity, blocking macrophage recruitment and activation, and utilizing nanoparticle-based drug delivery systems, may provide new avenues for combating HCC by modulating macrophage functions and tumor microenvironment dynamics.
Collapse
Affiliation(s)
- Shuairan Zhang
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Hang Dong
- Phase I Clinical Trials Center, The People's Hospital of China Medical University, Shenyang, PR China
| | - Xiuli Jin
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Jing Sun
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Yiling Li
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
18
|
Anwar AA, Jalan-Sakrikar N, Huebert RC. LncRNAs, RNA Therapeutics, and Emerging Technologies in Liver Pathobiology. Semin Liver Dis 2025; 45:1-14. [PMID: 39603269 DOI: 10.1055/a-2490-1921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The field of ribonucleic acid (RNA) biology has revealed an array of noncoding RNA species, particularly long noncoding RNAs (lncRNAs), which play crucial roles in liver disease pathogenesis. This review explores the diverse functions of lncRNAs in liver pathology, including metabolic-associated steatotic liver disease, hepatocellular carcinoma, alcohol-related liver disease, and cholangiopathies such as primary sclerosing cholangitis and cholangiocarcinoma. We highlight key lncRNAs that regulate lipid metabolism, inflammation, fibrosis, and oncogenesis in the liver, demonstrating their diagnostic and therapeutic potential. Emerging RNA-based therapies, such as mRNA therapy, RNA interference, and antisense oligonucleotides, offer approaches to modulate lncRNA activity and address liver disease at a molecular level. Advances in sequencing technologies and bioinformatics pipelines are simultaneously enabling the identification and functional characterization of novel lncRNAs, driving innovation in personalized medicine. In conclusion, this review highlights the potential of lncRNAs as biomarkers and therapeutic targets in liver disease and emphasizes the need for further research into their regulatory mechanisms and clinical applications.
Collapse
Affiliation(s)
- Abid A Anwar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
- Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Robert C Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
- Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| |
Collapse
|
19
|
Kamdem SD, Kamguia LM, Oumarou A, Bitye BMZ, Lennard K, Brombacher F, Spangenberg T, Demarta-Gatsi C, Nono JK. Reduced plasma levels of GM-CSF is a common feature of Schistosoma mansoni-infected school-aged children. Front Immunol 2025; 16:1474575. [PMID: 40092989 PMCID: PMC11906694 DOI: 10.3389/fimmu.2025.1474575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background Currently available schistosomiasis diagnostic and monitoring tools are limited, and the development of novel technologies is necessary to enhance disease diagnostic and surveillance by supporting elimination efforts. Novel disease-specific biomarkers can facilitate the development of these technologies. Through the comparison of parasite burden and host factors, we assessed whether host plasma cytokines could be used as robust biomarkers for intestinal schistosomiasis and associated pathology in school-aged children (SAC) living in endemic areas. Methods Levels of host plasma cytokines were measured in SAC from a low-to-moderate burden region five months deworming with praziquantel, using Luminex assay for exploration analysis and ELISA for validation. Results The concentration of GM-CSF, IL-2, and VEGF in plasma was significantly lower in schistosome-infected compared to non-infected children, as determined by Luminex assay. Further evaluation by ELISA revealed a negative correlation between GM-CSF plasma levels, but not those of IL-2 or VEGF, and S. mansoni egg burdens in infected individuals. Common coinfections in the study area such as geohelminths, hepatitis or malaria failed to alter plasma GM-CSF levels arguing in favor of a potential specific effect of S. mansoni infection on this cytokine. Receiver operating characteristic analysis confirmed GM-CSF as an acceptable predictive marker of S. mansoni infection, with an area under the curve (AUC) of 75%. Finally, the adjunct use of plasmatic GM-CSF thresholds for screening S. mansoni at-risk children and identify S. mansoni-infected ones increased the sensitivity of a single Kato-Katz test by averagely 15%. Conclusions Our findings highlight the potential of using plasma GM-CSF levels to biomark S. mansoni infection and improve the sensitivity of single Kato-Katz based diagnostic for low- to-moderate burden infections.
Collapse
Affiliation(s)
- Severin Donald Kamdem
- Unit of Immunobiology and Helminth Infections, Laboratory of Molecular Biology and Biotechnology, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Leonel Meyo Kamguia
- Unit of Immunobiology and Helminth Infections, Laboratory of Molecular Biology and Biotechnology, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
- Faculty of Health Sciences, Protestant University Institute of Yaoundé, Yaoundé, Cameroon
- Ecole Doctorale Regionale (EDR) d’Afrique Centrale en Infectiologie Tropicale, Université des Sciences et Techniques de Masuku (USTM), Franceville, Gabon
| | - Alim Oumarou
- District Hospital of Mfou, Ministry of Public Health, Yaoundé, Cameroon
| | - Bernard Marie Zambo Bitye
- Unit of Immunobiology and Helminth Infections, Laboratory of Molecular Biology and Biotechnology, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Katie Lennard
- Department of Integrated Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
- Immunology of Infectious Diseases Unit, South African Medical Research Centre, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Thomas Spangenberg
- Global Health R&D of Merck Healthcare, Ares Trading S.A., (a subsidiary of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Claudia Demarta-Gatsi
- Global Health R&D of Merck Healthcare, Ares Trading S.A., (a subsidiary of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Justin Komguep Nono
- Unit of Immunobiology and Helminth Infections, Laboratory of Molecular Biology and Biotechnology, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
- Division of Immunology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
20
|
Zhou Y, Ding Y, Li Y, Sheng Q, Han C, Fan Y, Wang Z, Lu B, Dou X, Zhang C. Sodium Butyrate Inhibits Necroptosis by Regulating MLKL via E2F1 in Intestinal Epithelial Cells of Liver Cirrhosis. J Clin Transl Hepatol 2025; 13:105-117. [PMID: 39917471 PMCID: PMC11797820 DOI: 10.14218/jcth.2024.00221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 02/09/2025] Open
Abstract
Background and Aims Necroptosis is critical for regulating intestinal epithelial cells (IECs). Butyric acid (BA), produced during intestinal microbial metabolism, protects the intestinal epithelial barrier. However, whether necroptosis occurs in IECs during liver cirrhosis and whether sodium butyrate (NaB) can regulate necroptosis have not yet been reported. In this study, we aimed to investigate whether IECs undergo necroptosis in cirrhosis and whether NaB can regulate necroptosis and the related regulatory mechanisms. Methods Serum levels of RIPK3, MLKL, and Zonulin, as well as fecal BA levels, were measured and correlated in 48 patients with liver cirrhosis and 20 healthy controls. A rat model of liver cirrhosis was established, and NaB was administered. The expressions of MLKL, p-MLKL, and tight junction proteins were measured. We conducted an in vitro investigation of the effect of NaB on necroptosis in the HT29 cell line. Results Serum levels of RIPK3, MLKL, and Zonulin in the liver cirrhosis group were higher, while fecal BA levels were lower than those in the control group. Zonulin levels were positively correlated with RIPK3 and MLKL levels, while fecal BA levels were negatively correlated with serum MLKL levels, but not with RIPK3 levels. NaB reduced the mRNA and protein expression of MLKL but had no effect on RIPK1 and RIPK3 in vitro. Rescue experiments demonstrated that NaB inhibited necroptosis through E2F1-mediated regulation of MLKL. Conclusions NaB alleviates intestinal mucosal injury and reduces necroptosis in IECs in liver cirrhosis. It also inhibits the necroptosis of IECs and protects the intestinal barrier by reducing E2F1 expression and downregulating MLKL expression levels. These results can be employed to develop a novel strategy for treating complications arising from liver cirrhosis.
Collapse
Affiliation(s)
- Yimeng Zhou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Viral Hepatitis, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Viral Hepatitis, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yanwei Li
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qiuju Sheng
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chao Han
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yaoxin Fan
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Viral Hepatitis, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ziyi Wang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bingchao Lu
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Viral Hepatitis, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Viral Hepatitis, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
21
|
Lin W, Chen Y, Lu M, Peng C, Chen X, Liu X, Wang Y. Identification and validation of neutrophil-related biomarkers in acute-on-chronic liver failure. Front Immunol 2025; 16:1477342. [PMID: 40070835 PMCID: PMC11893565 DOI: 10.3389/fimmu.2025.1477342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Dysfunction of peripheral blood neutrophils occurs in acute-on-chronic liver failure (ACLF). However, the molecular mechanisms of neutrophils involved in the pathophysiology of the ACLF remains poorly understood. Data downloaded from the GEO database (GSE142255) was used to identify both ACLF and neutrophil-related genes with the help of the limma package and Weighted Gene Co-Expression Network Analysis (WGCNA) algorithms. The analysis identified 288 ACLF-related differentially expressed genes (DEGs) in the circulating blood cells. Among these, three genes were found to be related to neutrophils and were identified as diagnostic genes, exhibiting high diagnostic efficacy as evidenced by an area under the curve (AUC) value of 1. Among these, matrix metallopeptidase-9 (MMP9) and S100 calcium binding protein A12 (S100A12) were upregulated, whereas C-C chemokine ligand 5 (CCL5) was downregulated in circulating immune cells from patients with ACLF compared to those from healthy controls. These findings were corroborated using an additional GEO dataset, GSE156382. The expression levels of the three key genes demonstrated a correlation with both ferroptosis and cuprotosis. Among the three diagnostic genes, only MMP9 was validated as differentially expressed through both quantitative real-time PCR (qRT-PCR) and western blot. Moreover, a significant elevation in plasma MMP9 levels was observed in patients with ACLF compared to those with chronic hepatitis B (CHB) and acute decompensated cirrhosis (AD). Notably, ACLF patients exhibiting elevated MMP9 levels (>175.8 ng/mL) experienced higher short-term mortality rates within both 30 and 90 days (p<0.001). In addition, a total of 21 drugs targeting the three diagnostic genes were identified from the Drug Bank database. Finally, the Kinase-TF-mRNA-miRNA network was constructed utilizing Cytoscape software. This study represents the initial application of WGCNA algorithms to identify novel biomarkers related to neutrophils in ACLF. Our findings offer new perspectives on the role of neutrophil in the pathogenesis of ACLF. However, additional research is required to substantiate the effects of these key genes and therapeutic agents on ACLF.
Collapse
Affiliation(s)
- Wei Lin
- Department of Orthopedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongping Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mingqin Lu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Peng
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiang Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoqin Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunyun Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
22
|
Fuster D, So-Armah K, Cheng DM, Blokhina E, Patts G, Lioznov D, Gnatienko N, Long MT, Freiberg MS, Tindle H, Samet JH. Is There an Association Between Cigarette Smoking and Advanced Liver Fibrosis in Smokers with HIV, Heavy Drinking and High Prevalence of HCV? J Clin Med 2025; 14:1169. [PMID: 40004700 PMCID: PMC11856725 DOI: 10.3390/jcm14041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Cigarette smoking has been associated with liver fibrosis in the setting of hepatitis C virus (HCV) infection but has not been studied among people with HIV (PWH) who consume alcohol. Methods: This is a cross-sectional study of PWH with heavy drinking and daily smoking in St. Petersburg, Russia. The primary independent variable was past 30-day cigarettes per day (cpd), and the secondary independent variable was pack-years at study entry. Advanced liver fibrosis was defined as FIB-4 > 3.25. Analyses were adjusted for gender, body mass index (BMI), past 30-day number of heavy drinking days, HCV and CD4 count. Results: Participants (n = 400) were two-thirds male (67.3%), young (median age 38 years), lean (median BMI 22), HCV antibody positive (84.5%) and not severely immune suppressed (median CD4 count 351). The median number of past-month cpd was 20 (IQR: 15-25), and the median pack-years was 24 (IQR: 17-31.8). The prevalence of advanced liver fibrosis was 11.3% (45/400). In the adjusted logistic regression analyses, we did not observe a significant association between cpd [middle (10.1-20 cigarettes) vs. lowest (5-10 cigarettes) category (adjusted odds ratio [aOR] (95% confidence interval [CI]): 1.06 (0.40-2.83), highest (>20.0 cigarettes) vs. lowest category aOR (95% CI): 0.65 (0.21-1.99), global p-value = 0.62]. The secondary analysis with pack-years yielded similar results [middle (20.1-30 pack-years) vs. lowest category (≤20 pack-years) aOR (95% CI): 0.81 (0.33-1.99), highest category (>30 pack-years) vs. lowest category aOR (95% CI): 0.91 (0.38-2.19); global p-value = 0.58]. Conclusions: In this Russian cohort of PWH, we did not detect an association between recent cigarette use or mean pack-years and advanced liver fibrosis.
Collapse
Affiliation(s)
- Daniel Fuster
- Addiction Unit, Internal Medicine Department, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Kaku So-Armah
- Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine/Boston Medical Center, Boston, MA 02118, USA; (K.S.-A.); (J.H.S.)
| | - Debbie M. Cheng
- Biostatistics Department, Boston University School of Public Health, Boston, MA 02118, USA;
| | - Elena Blokhina
- Laboratory of Clinical Psychopharmacology of Addictions, First Pavlov State Medical University, 197022 St. Petersburg, Russia;
| | - Gregory Patts
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA 02118, USA;
| | - Dmitry Lioznov
- Department of Infectious Diseases and Epidemiology, First Pavlov State Medical University, 197022 St. Petersburg, Russia;
- Smorodintsev Research Institute of Influenza, 197022 St. Petersburg, Russia
| | - Natalia Gnatienko
- Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Department of Medicine, Boston Medical Center, Boston, MA 02118, USA;
| | - Michelle T. Long
- Gastroenterology Section, Department of Medicine, Boston University School of Medicine/Boston Medical Center, Boston, MA 02118, USA;
| | - Matthew S. Freiberg
- Cardiovascular Medicine Division, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Hilary Tindle
- ViTAL, the Vanderbilt Center for Tobacco, Addiction and Lifestyle, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Jeffrey H. Samet
- Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine/Boston Medical Center, Boston, MA 02118, USA; (K.S.-A.); (J.H.S.)
- Department of Community Health Sciences, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
23
|
Park J, Lee YT, Agopian VG, Liu JS, Koltsova EK, You S, Zhu Y, Tseng HR, Yang JD. Liquid biopsy in hepatocellular carcinoma: Challenges, advances, and clinical implications. Clin Mol Hepatol 2025; 31:S255-S284. [PMID: 39604328 PMCID: PMC11925447 DOI: 10.3350/cmh.2024.0541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive primary liver malignancy often diagnosed at an advanced stage, resulting in a poor prognosis. Accurate risk stratification and early detection of HCC are critical unmet needs for improving outcomes. Several blood-based biomarkers and imaging tests are available for early detection, prediction, and monitoring of HCC. However, serum protein biomarkers such as alpha-fetoprotein have shown relatively low sensitivity, leading to inaccurate performance. Imaging studies also face limitations related to suboptimal accuracy, high cost, and limited implementation. Recently, liquid biopsy techniques have gained attention for addressing these unmet needs. Liquid biopsy is non-invasive and provides more objective readouts, requiring less reliance on healthcare professional's skills compared to imaging. Circulating tumor cells, cell-free DNA, and extracellular vesicles are targeted in liquid biopsies as novel biomarkers for HCC. Despite their potential, there are debates regarding the role of these novel biomarkers in the HCC care continuum. This review article aims to discuss the technical challenges, recent technical advancements, advantages and disadvantages of these liquid biopsies, as well as their current clinical application and future directions of liquid biopsy in HCC.
Collapse
Affiliation(s)
- Jaeho Park
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yi-Te Lee
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Vatche G. Agopian
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Jessica S Liu
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Ekaterina K. Koltsova
- Smidt Heart Institute, Department of Medicine, Department of Biomedical Sciences, 8700 Beverly Blvd, Los Angeles, CA, USA
| | - Sungyong You
- Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
24
|
Lin YD, Li XY, Shao LW, Liu AJ. Methylation of SOX1 and PAX1 Are Risk Factors and Potential Biomarkers for Cervical Lesions. World J Oncol 2025; 16:104-112. [PMID: 39850527 PMCID: PMC11750758 DOI: 10.14740/wjon1985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/10/2024] [Indexed: 01/25/2025] Open
Abstract
Background The correlation between methylation of paired box gene 1 (PAX1) and sex determining region Y-box 1 (SOX1) with human papillomavirus (HPV) infection and the progression of cervical lesions is not well understood. This study aims to explore the potential value of PAX1 and SOX1 as diagnostic biomarkers for cervical diseases. Methods A total of 139 cervical biopsy tissue samples were obtained from the Department of Pathology, the Seventh Medical Center, Chinese PLA General Hospital from 2021 to 2023. The samples include 32 cases of chronic cervicitis (inflammation group), 30 cases of low-grade squamous intraepithelial lesions (LSIL group), 50 cases of high-grade squamous intraepithelial lesions (HSIL group), and 27 cases of cervical squamous cell carcinoma (CSCC group). DNA was extracted from paraffin-embedded tissues, and the levels of HPV infection and methylation of PAX1 and SOX1 were detected. Results The methylation index (M-index) of PAX1 and SOX1 in the HSIL and CSCC groups is significantly higher than in the inflammation group (both P < 0.0001), with no significant difference between the LSIL and inflammation groups. There is no significant difference in the positive PAX1 and SOX1 methylation rate with HPV infection and age. The positive rates of PAX1 methylation in the inflammation, LSIL, HSIL, and CSCC groups were 3.13%, 10.00%, 44.00%, and 88.89%, respectively. The positive rates of SOX1 methylation were 3.13%, 10.00%, 40.00%, and 77.78%, respectively, and increasing with the progression of cervical lesions (R2 = 0.9189/R2 = 0.9279, P < 0.0001/P < 0.0001). Comparing LSIL, HSIL, and CSCC with the inflammation group and using cervical biopsy pathology diagnosis as the gold standard, methylation of PAX1 and SOX1 is a risk factor for HSIL and CSCC, with odds ratio (OR) values significantly increasing as lesions progress. The sensitivity of PAX1 and SOX1 methylation to cervical lesions increases with the progression of the lesions. Conclusions Methylation of SOX1 and PAX1 is not associated with HPV infection. The positive rate of methylation for SOX1 and PAX1 is positively correlated with cervical lesions, which can serve as potential biomarkers for HSIL and CSCC. They are risk factors and potential screening indicators for HSIL and above cervical lesions.
Collapse
Affiliation(s)
- Yan Die Lin
- Department of Pathology, The Seventh Medical Center, Chinese PLA General Hospital, Beijing 100700, China
- These authors contributed equally to this work
| | - Xiao Yue Li
- Department of Pathology, The Seventh Medical Center, Chinese PLA General Hospital, Beijing 100700, China
- These authors contributed equally to this work
| | - Li Wei Shao
- Department of Pathology, The Seventh Medical Center, Chinese PLA General Hospital, Beijing 100700, China
| | - Ai Jun Liu
- Department of Pathology, The Seventh Medical Center, Chinese PLA General Hospital, Beijing 100700, China
| |
Collapse
|
25
|
Tan X, Xun L, Yin Q, Chen C, Zhang T, Shen T. Epigenetic Modifications in HBV-Related Hepatocellular Carcinoma. J Viral Hepat 2025; 32:e14044. [PMID: 39868653 DOI: 10.1111/jvh.14044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/13/2024] [Accepted: 11/30/2024] [Indexed: 01/28/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Hepatitis B virus (HBV) is the main pathogen for HCC development. HBV covalently closed circular DNA (cccDNA) forms extra-host chromatin-like minichromosomes in the nucleus of hepatocytes with host histones, non-histones, HBV X protein (HBx) and HBV core protein (HBc). Epigenetic alterations are dynamic and reversible, which regulate gene expression without altering the DNA sequence and play a pivotal role in the regulation of HCC onset and progression. The aim of this review is to elucidate the deregulation of epigenetic mechanisms involved in the pathogenesis of HBV-related HCC (HBV-HCC), including post-translational histone and non-histone modifications, DNA hypermethylation and hypomethylation, non-coding RNA modification on HBV cccDNA minichromosomes and host factors, effecting the replication/transcription of HBV cccDNA and transcription/translation of host genes, and thus HBV-HCC progression. It is expected that the epigenetic regulation perspective provides new ways for more in-depth development of therapeutic control of HBV-HCC.
Collapse
Affiliation(s)
- Xiaoqing Tan
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| | - Linting Xun
- Department of Gastroenterology, the First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People's Republic of China
| | - Qi Yin
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China, China
| | - Chaohui Chen
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Tao Zhang
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| | - Tao Shen
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| |
Collapse
|
26
|
Wang JL, Jiang SW, Hu AR, Shi XJ, Zhou AW, Lin K, Fan Y, Jin MH, Zhang HJ. A model based on chitinase 3-like protein for expecting liver severity of hepatitis B virus infections in the immune tolerance phase. Clin Chim Acta 2025; 567:120085. [PMID: 39667422 DOI: 10.1016/j.cca.2024.120085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND The question of whether to treat patients with chronic hepatitis B (CHB) during the immune tolerance (IT) period is a matter of ongoing debate, as it is difficult to discern different levels of liver disease severity. We created and assessed a novel diagnostic model for identifying significant liver tissue damage in individuals with CHB in IT phase. METHODS From November 2018 to December 2022, a cross-sectional study of 311 patients with chronic hepatitis B virus infection (HBV DNA > 30 IU/mL) at Ningbo No. 2 Hospital, Ningbo, China, who underwent liver biopsy, including 44 patients in IT phase. Utilizing univariate regression analyses and logistics analysis, and model was developed and validated to predict the severity of hepatic inflammatory and fibrosis in CHB patients and in IT phase. RESULTS Chitinase 3-like Protein (CHI3L1), albumin (ALB), alanine transaminase (ALT) / aspartate aminotransferase (AST) were identified as independent predictors of liver lesion severity in CHB patients with IT. The three were combined to build the model (named as CAA index), which demonstrated good performance. The CAA index achieved an area under the receiver operating characteristic curve (AUC) of 0.916 (95 % CI, 0.820-1.000) and AUC of validation group was 0.875 (95 % CI, 0.683-1.000). CONCLUSIONS CHI3L1 serves as an independent measure of liver fibrosis and inflammation in CHB. This diagnostic model has some value in assessing the severity of the patient's liver lesion severity and may be a reliable non-invasive diagnostic model helping determine whether treatment is necessary among CHB patients in IT phase.
Collapse
Affiliation(s)
- Jia-Lan Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China; Liver Diseases Center, Ningbo No. 2 Hospital, Ningbo 315020, Zhejiang Province, China
| | - Su-Wen Jiang
- Liver Diseases Center, Ningbo No. 2 Hospital, Ningbo 315020, Zhejiang Province, China
| | - Ai-Rong Hu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China; Liver Diseases Center, Ningbo No. 2 Hospital, Ningbo 315020, Zhejiang Province, China.
| | - Xiao-Jun Shi
- Liver Diseases Center, Ningbo No. 2 Hospital, Ningbo 315020, Zhejiang Province, China
| | - Ai-Wu Zhou
- Liver Diseases Center, Ningbo No. 2 Hospital, Ningbo 315020, Zhejiang Province, China
| | - Ken Lin
- Ningbo University Health Science Center, Ningbo 315211, Zhejiang Province, China
| | - Ying Fan
- School of Medicine, Shaoxing University, Shaoxing 31200, Zhejiang Province, China
| | - Meng-Han Jin
- Ningbo University Health Science Center, Ningbo 315211, Zhejiang Province, China
| | - Hao-Jin Zhang
- School of Medicine, Shaoxing University, Shaoxing 31200, Zhejiang Province, China
| |
Collapse
|
27
|
Wang D, Yu X, Yang Y. Investigating SNHG3 as a potential therapeutic approach for HCC stem cells. Gene 2025; 935:149022. [PMID: 39427830 DOI: 10.1016/j.gene.2024.149022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/13/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Hepatocellular Carcinoma (HCC) is a common malignant tumor worldwide. Long Non-Coding RNA (lncRNA) has gained attention in tumor biology, and this study aims to investigate the role of lncRNA SNHG3 in HCC, specifically in the self-renewal and maintenance of liver cancer stem cells. METHODS The expression of lncRNA SNHG3 was analyzed in HCC and adjacent normal tissue using the TCGA database. The expression levels of SNHG3 in HCC cell lines (Hep3B, HepG2, Huh7) were detected using qRT-PCR and Western blot techniques. Functional assays, including CCK-8, soft agar colony formation, and tumor sphere formation, were performed to evaluate the impact of SNHG3 on HCC stem cell functionality. MeRIP-qPCR was also used to investigate the regulatory role of SNHG3 in m6A modification of ITGA6 mRNA mediated by METTL3. RESULTS The study found that SNHG3 was significantly upregulated in HCC tissue and cell lines compared to normal liver tissue. SNHG3 expression correlated with the pathological stage, metastasis status, and tumor size of liver cancer. Inhibiting SNHG3 reduced proliferation, colony formation, and tumor sphere formation ability in HCC stem cells. SNHG3 also played a role in regulating the m6A modification and expression of ITGA6 through METTL3. CONCLUSION This study emphasizes the upregulation of lncRNA SNHG3 and its role in HCC stem cell self-renewal. SNHG3 may regulate the m6A modification of ITGA6 mRNA through its interaction with METTL3, impacting the function of liver cancer stem cells. These findings support the potential of targeting SNHG3 as a therapeutic approach for HCC.
Collapse
Affiliation(s)
- Dingmao Wang
- Department of Hepatobiliary Surgery, Haikou People's Hospital, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, PR China
| | - Xiao Yu
- The 2nd Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| | - Yijun Yang
- Department of Hepatobiliary Surgery, Haikou People's Hospital, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, PR China.
| |
Collapse
|
28
|
Antwi SO, Jnr. Siaw AD, Armasu SM, Frank JA, Yan IK, Ahmed FY, Izquierdo-Sanchez L, Boix L, Rojas A, Banales JM, Reig M, Stål P, Gómez MR, Wangensteen KJ, Singal AG, Roberts LR, Patel T. Genome-Wide DNA Methylation Markers Associated With Metabolic Liver Cancer. GASTRO HEP ADVANCES 2025; 4:100621. [PMID: 40275933 PMCID: PMC12019016 DOI: 10.1016/j.gastha.2025.100621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/17/2025] [Indexed: 04/26/2025]
Abstract
Background and Aims Metabolic liver disease is the fastest-rising cause of hepatocellular carcinoma (HCC), but the underlying molecular processes that drive HCC development in the setting of metabolic perturbations are unclear. We investigated the role of aberrant DNA methylation in metabolic HCC development in a multicenter international study. Methods We used a case-control design, frequency-matched on age, sex, and study site. Genome-wide profiling of peripheral blood leukocyte DNA was performed using the 850k EPIC array. The study sample was split 80% and 20% for training and validation. Cell type proportions were estimated from the methylation data. Differential methylation analysis was performed adjusting for cell type, generating area under the receiver-operating characteristic curves (AUC-ROC). Results We enrolled 272 metabolic HCC patients and 316 control patients with metabolic liver disease from 6 sites. Fifty-five differentially methylated CpGs were identified; 33 hypermethylated and 22 hypomethylated in cases vs controls. The panel of 55 CpGs discriminated between the cases and controls with AUC = 0.79 (95% confidence interval [CI] = 0.71-0.87), sensitivity = 0.77 (95% CI = 0.66-0.89), and specificity = 0.74 (95% CI = 0.64-0.85). The 55-CpG classifier panel performed better than a base model that comprised age, sex, race, and diabetes mellitus (AUC = 0.65, 95% CI = 0.55-0.75; sensitivity = 0.62, 95% CI = 0.49-0.75; and specificity = 0.64, 95% CI = 0.52-0.75). A multifactorial model that combined the 55 CpGs with age, sex, race, and diabetes yielded AUC = 0.78 (95% CI = 0.70-0.86), sensitivity = 0.81 (95% CI = 0.71-0.92), and specificity = 0.67 (95% CI = 0.55-0.78). Conclusion A panel of 55 blood leukocyte DNA methylation markers differentiates patients with metabolic HCC from control patients with benign metabolic liver disease, with a slightly higher sensitivity when combined with demographic and clinical information.
Collapse
Affiliation(s)
- Samuel O. Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida
| | - Ampem Darko Jnr. Siaw
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Sebastian M. Armasu
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Jacob A. Frank
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Irene K. Yan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Fowsiyo Y. Ahmed
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Loreto Boix
- BCLC Group, Liver Unit, ICMDM, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona. Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD); Barcelona University, Barcelona, Spain
| | - Angela Rojas
- SeLiver Group, UCM Digestive Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Maria Reig
- BCLC Group, Liver Unit, ICMDM, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona. Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD); Barcelona University, Barcelona, Spain
| | - Per Stål
- Department of Upper GI Diseases, Karolinska University Hospital, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Romero Gómez
- SeLiver Group, UCM Digestive Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Kirk J. Wangensteen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Amit G. Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Tushar Patel
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
29
|
Shahidin, Wang Y, Wu Y, Chen T, Wu X, Yuan W, Zhu Q, Wang X, Zi C. Selenium and Selenoproteins: Mechanisms, Health Functions, and Emerging Applications. Molecules 2025; 30:437. [PMID: 39942544 PMCID: PMC11820089 DOI: 10.3390/molecules30030437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Selenium (Se) is an essential trace element crucial for human health that primarily functions as an immunonutrient. It is incorporated into polypeptides such as selenocysteine (SeC) and selenomethionine (SeMet), two key amino acids involved in various biochemical processes. All living organisms can convert inorganic Se into biologically active organic forms, with SeMet being the predominant form and a precursor for SeC production in humans and animals. The human genome encodes 25 selenoprotein genes, which incorporate low-molecular-weight Se compounds in the form of SeC. Organic Se, especially in the form of selenoproteins, is more efficiently absorbed than inorganic Se, driving the demand for selenoprotein-based health products, such as functional foods. Se-enriched functional foods offer a practical means of delivering bioavailable Se and are associated with enhanced antioxidant properties and various health benefits. Recent advancements in selenoprotein synthesis have improved our understanding of their roles in antioxidant defense, cancer prevention, immune regulation, anti-inflammation, hypoglycemia, cardiovascular health, Alzheimer's disease, fertility, and COVID-19. This review highlights key selenoproteins and their biological functions, biosynthetic pathways, and emerging applications while highlighting the need for further research.
Collapse
Affiliation(s)
- Shahidin
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Yan Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Yilong Wu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Taixia Chen
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Xiaoyun Wu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Wenjuan Yuan
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Qiangqiang Zhu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
| | - Xuanjun Wang
- College of Resources, Environment, and Chemistry, Chuxiong Normal University, No. 546 S Rd. Lucheng, Chuxiong 675099, China
| | - Chengting Zi
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (S.); (Y.W.); (Y.W.); (T.C.); (X.W.); (W.Y.); (Q.Z.)
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
30
|
Lim H, El-Serag HB, Luster M, Grove ML, Byun J, Jung Y, Han Y, Boerwinkle E, Amos CI, Thrift AP. DNA Methylation Profile in Buffy Coat Identifies Methylation Differences Between Cirrhosis with and Without Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:266. [PMID: 39858049 PMCID: PMC11763440 DOI: 10.3390/cancers17020266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cirrhosis is the precursor to most cases of hepatocellular carcinoma (HCC). Understanding the mechanisms leading to the transition from cirrhosis to HCC and identifying key biomarkers is crucial to developing effective screening strategies and reducing HCC-related mortality. DNA methylation is associated with gene inactivation and plays an important role in physiological and pathological processes; however, its role in cirrhosis progression to HCC is unknown. METHODS We performed genome-wide DNA methylation profiling using Illumina Infinium MethylationEPI BeadChip in pre-diagnostic samples from 22 cirrhosis patients who subsequently developed HCC and 22 cirrhosis patients who remained HCC-free during an average 4-year follow-up. In a secondary analysis, we examined a subset of patients without hepatitis C virus (HCV) infection. RESULTS We identified three differentially methylated positions (DMPs) located in ADAM12 (cg13674437) and PSD3 (cg06758847 and cg24595678) that show a strong association with HCC risk (lower median vs. higher median hazards ratio (HR): HR cg13674437 = 0.34, 95% CI = 0.14-0.83; HR cg06758847 = 4.89, 95% CI = 1.79-13.33; HR cg24595678 = 11.19, 95% CI = 3.27-38.35). After excluding all HCV-active patients from our analysis, the HR for the DMPs remained significant. CONCLUSIONS In conclusion, the findings in this study support the theory that buffy coat-derived DNA methylation markers could be used to identify biomarkers among cirrhosis patients at high risk for HCC before clinical symptoms appear. A further study with a large prospective cohort is required to validate these findings.
Collapse
Affiliation(s)
- Hyeyeun Lim
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (H.B.E.-S.); (M.L.)
| | - Michelle Luster
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (H.B.E.-S.); (M.L.)
| | - Megan L. Grove
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.L.G.); (E.B.)
| | - Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA; (J.B.); (Y.H.)
| | - Yuri Jung
- Ridgewood High School, Ridgewood, NJ 07450, USA;
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA; (J.B.); (Y.H.)
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.L.G.); (E.B.)
| | - Christopher I. Amos
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA; (J.B.); (Y.H.)
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77054, USA
| | - Aaron P. Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77054, USA
| |
Collapse
|
31
|
Trombetti S, Cimbalo A, Grosso M, Vila-Donat P, Mañes J, Manyes L. Proteomic Analysis of the Murine Liver Response to Oral Exposure to Aflatoxin B1 and Ochratoxin A: The Protective Role to Bioactive Compounds. Toxins (Basel) 2025; 17:29. [PMID: 39852982 PMCID: PMC11768807 DOI: 10.3390/toxins17010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Aflatoxin B1 (AFB1) and Ochratoxin A (OTA) are considered the most important mycotoxins in terms of food safety. The aim of this study was to evaluate the hepatotoxicity of AFB1 and OTA exposure in Wistar rats and to assess the beneficial effect of fermented whey (FW) and pumpkin (P) as functional ingredients through a proteomic approach. For the experimental procedures, rats were fed AFB1 and OTA individually or in combination, with the addition of FW or a FW-P mixture during 28 days. For proteomics analysis, peptides were separated using a LC-MS/MS-QTOF system and differentially expressed proteins (DEPs) were statistically filtered (p < 0.05) distinguishing males from females. Gene ontology visualization allowed the identification of proteins involved in important biological processes such as the response to xenobiotic stimuli and liver development. Likewise, KEGG pathway analysis reported the metabolic routes as the most affected, followed by carbon metabolism and biosynthesis of amino acids. Overall, the results highlighted a strong downregulation of DEPs in the presence of AFB1 and OTA individually but not with the mixture of both, suggesting a synergistic effect. However, FW and P have helped in the mitigation of processes triggered by mycotoxins.
Collapse
Affiliation(s)
- Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (S.T.); (M.G.)
| | - Alessandra Cimbalo
- Biotech Agrifood, Faculty of Pharmacy and Food Sciences, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (A.C.); (J.M.); (L.M.)
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (S.T.); (M.G.)
| | - Pilar Vila-Donat
- Biotech Agrifood, Faculty of Pharmacy and Food Sciences, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (A.C.); (J.M.); (L.M.)
| | - Jordi Mañes
- Biotech Agrifood, Faculty of Pharmacy and Food Sciences, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (A.C.); (J.M.); (L.M.)
| | - Lara Manyes
- Biotech Agrifood, Faculty of Pharmacy and Food Sciences, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (A.C.); (J.M.); (L.M.)
| |
Collapse
|
32
|
Xu M, Xiao H, Zou X, Pan L, Song Q, Hou L, Zeng Y, Han Y, Zhou Z. Mechanisms of levan in ameliorating hyperuricemia: Insight into levan on serum metabolites, gut microbiota, and function in hyperuricemia rats. Carbohydr Polym 2025; 347:122665. [PMID: 39486924 DOI: 10.1016/j.carbpol.2024.122665] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 11/04/2024]
Abstract
This study aims to investigate the effects of levan on the progression of hyperuricemia (HUA) rats and elucidate its underlying mechanisms. After levan intervention, both low and high-dose groups exhibited a significant decrease in serum uric acid (UA) levels, reaching 71.0 % and 77.5 %, respectively, compared to the model group. Furthermore, levan could alleviate renal pathological damage caused by glomerular cell vacuolation, inflammatory infiltration and collagen deposition. The results of enzyme activity assay and real-time fluorescence quantitative PCR showed that levan decreased UA production by inhibiting adenosine deaminase (ADA) activity and gene expression in liver; it upregulated ATP-binding cassette subfamily G member 2 protein (ABCG2) and organic anion transporter 1 (OAT1) transporter gene expression in the kidney, promoting UA excretion. Gut microbiome analysis indicated that levan regulated gut flora dysbiosis induced by HUA, resulting in up-regulated the abundance of beneficial bacteria (Muribaculaceae, Faecalibaculum, Bifidobacterium, and Lactobacillus) and decreased conditioned pathogenic bacteria (Escherichia_Shigella and Proteus). Non-targeted metabolomics showed changes in various serum metabolites associated with glycerophospholipid metabolism, lipid metabolism, and inflammation following oral administration of levan. Therefore, levan may be a promising functional dietary supplement for regulating the gut flora and remodeling of metabolic disorders in individuals with HUA.
Collapse
Affiliation(s)
- Min Xu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Huazhi Xiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Xuan Zou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Lei Pan
- School of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qiaozhi Song
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Luying Hou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Yihong Zeng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Ye Han
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Zhijiang Zhou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| |
Collapse
|
33
|
Zhu W, Fan C, Liu B, Qin J, Fan A, Yang Z, Zhang H, Zhou W. Therapeutic targets for hepatocellular carcinoma identified using proteomics and Mendelian randomization. J Gastroenterol Hepatol 2025; 40:282-293. [PMID: 39477889 DOI: 10.1111/jgh.16785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND AND AIM Hepatocellular carcinoma (HCC) emerges as a formidable malignancy marked by elevated morbidity and mortality rates, coupled with a dismal prognosis. The revelation of gene-protein associations has presented an avenue for the exploration of novel therapeutic targets. METHODS Pooling plasma proteomic data (seven published GWAS) and HCC data (DeCODE cohort), we applied MR to identify potential drug targets, which were further validated in the FinnGen cohort and UK Biobank. Subsequent colocalization and summary-data-based Mendelian randomization analyses were performed for potential associations of this set of proteins. In addition, enrichment information pathways were investigated in depth by KEGG pathway analysis, single-cell sequencing, PPI and DGIdb, ChEMBL, and DrugBank database analyses, specific cell types enriched for expression were identified, interacting proteins were identified, and finally, druggability was assessed. RESULTS In summary, the levels of 10 proteins are linked to HCC risk. Elevated levels of TFPI2 as well as decreased levels of ALDH1A1, KRT18, ADAMTS13, TIMD4, SCLY, HRSP12, TNFAIP6, FTCD, and DDC are associated with increased HCC risk. Notably, HRSP12 show the strongest evidence. These genes are primarily expressed in specific cell types within the HCC TME. Moreover, intricate protein-protein interactions, involving key players like ALDH1A1 and RIDA, ALDH1A1 and DDC, and ALDH1A1 and KRT18, contribute significantly to the amino acid metabolism and dopaminergic neurogenesis pathway. Proteins such as ALDH1A1, KRT18, TFPI2, and DDC are promising targets for HCC therapy and broader cancer drug development. Targeting these proteins offers substantial potential in advancing HCC treatment strategies. CONCLUSIONS This research delineates 10 protein biomarkers linked to HCC risk and offers novel perspectives on its etiology, as well as promising avenues for the screening of HCC protein markers and therapeutic agents.
Collapse
Affiliation(s)
- Weixiong Zhu
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Chuanlei Fan
- Nanchang Central Hospital, Jiangxi Provincial University of Traditional Chinese Medicine, nanchang, 330000, China
| | - Bo Liu
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jianqi Qin
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Aodong Fan
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Zengxi Yang
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Hui Zhang
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
34
|
Li P, Liu S, Wang T, Wang F, Li J, Qi Q, Zhang S, Xie Y, Li J, Zhu Y, Yang S, Yin G, He X, Li S, Xu H, Xiong M, Li G, Zhang Y, Du L, Wang C. Multisite DNA methylation alterations of peripheral blood mononuclear cells serve as novel biomarkers for the diagnosis of AIS/stage I lung adenocarcinoma: a multicenter cohort study. Int J Surg 2025; 111:40-54. [PMID: 39352118 PMCID: PMC11745624 DOI: 10.1097/js9.0000000000002101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Early diagnosis remains an obstacle for improving the outcome of lung adenocarcinoma (LUAD). DNA methylation changes in peripheral blood mononuclear cells (PBMCs) could reflect an immune response to tumorigenesis, providing the theoretical basis for early cancer diagnosis based on immune cell profiling. METHODS This multi-center study evaluated the DNA methylation patterns based on PBMCs samples from 1115 individuals at nine medical centers. Genome-wide DNA methylation profiling of PBMCs in a discovery cohort (35 LUAD patients and 50 healthy controls) was performed using Illumina 850K microarray. Candidate differentially methylated CpG positions (DMPs) were selected and validated in a two-step DMPs screening cohort (65 LUAD patients and 80 healthy controls) by pyrosequencing and multiple target region methylation enrichment sequencing (MTRMES). Then, an early LUAD Diagnostic Panel (LDP score) based on multisite methylation-specific chip-based digital PCR was constructed in a training set and then confirmed in a validation set from the LDP score development cohort (389 AIS/stage I LUAD patients and 293 healthy controls). Besides, we included 157 other cancer patients, including 52 gastric cancer (GC) patients, 50 breast cancer (BC) patients, and 55 colorectal cancer (CRC) patients to assess the specificity of the LDP score. In addition, we also evaluated the early warning ability of LDP score for LUAD in a prospective cohort (46 people who were at high-risk of developing LC). RESULTS A total of 1415 LUAD-specific DMPs were identified. Then, six DMPs were selected for validation and three DMPs were finally verified. The LDP score was constructed by combining the three DMPs, age, and sex, and showed an AUC of 0.916, sensitivity of 88.17%, and specificity of 80.20% in a combined set, outperforming traditional methods, such as CEA and CT (detection rate: 87.79% vs. 4.69%; 87.79% vs. 35.21%). This diagnostic performance was confirmed in sub-types of LUAD with clinical challenges, such as 6-20 mm LUAD (AUC: 0.914, 95% CI: 0.889-0.934) and ground-glass nodules (AUC: 0.916, 95% CI: 0.889-0.938). Importantly, our LDP score had significant improvement in terms of selecting high-risk individuals who should receive low-dose computed tomography (87.80% vs. 9.28%). Remarkably, the LDP score could predict LUAD around 2 years before clinical diagnosis in our prospective cohort. CONCLUSIONS The novel developed LDP score represented a convenient and effective assay for the detection of AIS/stage I LUAD with high sensitivity and specificity, and had demonstrated unique advantages over traditional detection methods.
Collapse
Affiliation(s)
- Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Shibiao Liu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Tiantian Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, People’s Republic of China
| | - Fang Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Qiuchen Qi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yan Xie
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Jianping Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yongcai Zhu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Suli Yang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Guotao Yin
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Xiaoyi He
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Department of Radiology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Shijun Li
- Department of Clinical Laboratory, The First Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Huiting Xu
- Departmemt of Clinical Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Jiangsu, People’s Republic of China; Medical School of Nantong University, Nantong, People’s Republic of China
| | - Mengqiu Xiong
- Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Guanghua Li
- Department of Clinical Laboratory, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Yi Zhang
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, People’s Republic of China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, People’s Republic of China
| |
Collapse
|
35
|
Solhi R, Pourhamzeh M, Zarrabi A, Hassan M, Mirzaei H, Vosough M. Novel biomarkers for monitoring and management of hepatocellular carcinoma. Cancer Cell Int 2024; 24:428. [PMID: 39719624 DOI: 10.1186/s12935-024-03600-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024] Open
Abstract
Due to current challenges in the early detection, less than 40% of individuals diagnosed with hepatocellular carcinoma (HCC) are viable candidates for surgical intervention. Therefore, validating and launching of a novel precise diagnostic approach is essential for early diagnosis. Based on developing evidence using circulating tumor cells and their derivatives, circulating miRNAs, and extracellular vesicles (EVs), liquid biopsy may offer a reliable platform for the HCC's early diagnosis. Each liquid biopsy analyte may provide significant areas for diagnosis, prognostic assessment, and treatment monitoring of HCC patients depending on its kind, sensitivity, and specificity. The current review addresses potential clinical applications, current research, and future developments for liquid biopsy in HCC management.
Collapse
Affiliation(s)
- Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Departments of Pathology and Medicine, UC San Diego, La Jolla, CA, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
36
|
Li X, Liu P, Wang Z, Wei X, Gao S, Fan Y, Liu H, Wang K. The value of promoter methylation of fibroblast factor 21 (FGF21) in predicting the course of chronic hepatitis B and the occurrence of oxidative stress. Virol J 2024; 21:332. [PMID: 39710689 PMCID: PMC11664819 DOI: 10.1186/s12985-024-02605-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Oxidative stress plays a crucial role in the pathogenesis of HBV. This study aimed to investigate the value of fibroblast growth factor 21 (FGF21) promoter methylation in the occurrence and development of chronic hepatitis B (CHB) oxidative stress. METHODS A total of 241 participants including 221 patients with CHB and 20 healthy controls (HCs) were recruited. Methylation level of FGF21 promoter in peripheral blood mononuclear cells was quantitatively determined. Enzyme-linked immunosorbent assay was used to assess oxidative stress in CHB patients. RESULTS Our study shows that the FGF21 methylation level was significantly lower in HBeAg-positive CHB patients compared to HBeAg-negative CHB patients and HCs (P < 0.0001). The oxidative stress of HBeAg-positive CHB patients was more severe. Further correlation analysis showed that there was a significant correlation between the methylation level of FGF21 promoter and the occurrence of oxidative stress in CHB patients. In addition, assessment based on FGF21 promoter methylation level proved effective for predicting oxidative stress occurrence and disease progression among CHB patients. CONCLUSION FGF21 promoter methylation level is an important marker for predicting oxidative stress and disease progression in patients with CHB.
Collapse
Affiliation(s)
- Xue Li
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China
| | - Pei Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China
| | - Zhaohui Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China
| | - Xuefei Wei
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China
| | - YuChen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China
| | - Huihui Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China.
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, China.
- Institute of Hepatology, Shandong University, Jinan, 250012, China.
| |
Collapse
|
37
|
Wojtukiewicz MZ, Mysliwiec M, Tokajuk A, Kruszewska J, Politynska B, Jamroze A, Wojtukiewicz AM, Tang DG, Honn KV. Tissue factor pathway inhibitor-2 (TFPI-2)-an underappreciated partaker in cancer and metastasis. Cancer Metastasis Rev 2024; 43:1185-1204. [PMID: 39153052 PMCID: PMC11554837 DOI: 10.1007/s10555-024-10205-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
The coagulation system is known to play an important role in cancer development and metastasis, but the precise mechanisms by which it does so remain incompletely understood. With this in mind, we provide an updated overview of the effects of TFPI-2, a protease inhibitor, on cancer development and metastasis. TFPI-2 interacts with the thrombin cascade and also employs other mechanisms to suppress cancer growth and dissemination, which include extracellular matrix stabilization, promotion of caspase-mediated cell apoptosis, inhibition of angiogenesis and transduction of intracellular signals. Down-regulation of TFPI-2 expression is well documented in numerous types of neoplasms, mainly via promoter methylation. However, the exact role of TFPI-2 in cancer progression and possible approaches to up-regulate TFPI-2 expression warrant further studies. Strategies to reactivate TFPI-2 may represent a promising direction for future anticancer studies and therapy development.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland.
- Department of Clinical Oncology, Comprehensive Cancer Center of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland.
| | - Marta Mysliwiec
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Anna Tokajuk
- Department of Clinical Oncology, Comprehensive Cancer Center of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Joanna Kruszewska
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Barbara Politynska
- Department of Psychology and Philosophy, Medical University of Bialystok, 37 Szpitalna, 15-295, Bialystok, Poland
- Robinson College, University of Cambridge, Grange Road, Cambridge, CB3 9AN, UK
| | - Anmbreen Jamroze
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Anna M Wojtukiewicz
- Department of Psychology and Philosophy, Medical University of Bialystok, 37 Szpitalna, 15-295, Bialystok, Poland
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kenneth V Honn
- Department of Pathology-School of Medicine, Bioactive Lipids Research Program, Wayne State University, 540 East Canfield Avenue, Detroit, MI, 48201, USA
- Karmanos Cancer Institute, 4100 John R St, Detroit, MI, 48201, USA
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, MI, 48202, USA
| |
Collapse
|
38
|
Komiya Y, Kamiya M, Oba S, Kawata D, Iwai H, Shintaku H, Suzuki Y, Miyamoto S, Tobiume M, Kanno T, Ainai A, Suzuki T, Hasegawa H, Hosoya T, Yasuda S. Necroptosis in alveolar epithelial cells drives lung inflammation and injury caused by SARS-CoV-2 infection. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167472. [PMID: 39154794 DOI: 10.1016/j.bbadis.2024.167472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
COVID-19, caused by SARS-CoV-2 infection, results in irreversible or fatal lung injury. We assumed that necroptosis of virus-infected alveolar epithelial cells (AEC) could promote local inflammation and further lung injury in COVID-19. Since CD8+ lymphocytes induced AEC cell death via cytotoxic molecules such as FAS ligands, we examined the involvement of FAS-mediated cell death in COVID-19 patients and murine COVID-19 model. We identified the occurrence of necroptosis and subsequent release of HMGB1 in the admitted patients with COVID-19. In the mouse model of COVID-19, lung inflammation and injury were attenuated in Fas-deficient mice compared to Fas-intact mice. The infection enhanced Type I interferon-inducible genes in both groups, while inflammasome-associated genes were specifically upregulated in Fas-intact mice. The treatment with necroptosis inhibitor, Nec1s, improved survival rate, lung injury, and systemic inflammation. SARS-CoV-2 induced necroptosis causes cytokine induction and lung damage, and its inhibition could be a novel therapeutic strategy for COVID-19.
Collapse
Affiliation(s)
- Yoji Komiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Mari Kamiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Seiya Oba
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Daisuke Kawata
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Hiroshi Shintaku
- Division of Pathology, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Yoshio Suzuki
- Department of Clinical Pathology, Asahi General Hospital, I-1326, Asahi, Chiba 289-2511, Japan
| | - Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Minoru Tobiume
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Hideki Hasegawa
- WHO Collaborating Centre for Reference and Research on Influenza, Tokyo, Japan; Research Center for Influenza and Respiratory Virus, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan.
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| |
Collapse
|
39
|
Xiang P, Li P, Yuan X, Zhao X, Xiao Z, Chen B, Liu K, Bischof E, Han J. Exon 1 methylation status of CDH13 is associated with decreased overall survival and distant metastasis in patients with postoperative colorectal cancer. Discov Oncol 2024; 15:725. [PMID: 39611998 DOI: 10.1007/s12672-024-01604-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Cadherin 13 (CDH13) is a member of the cadherin superfamily that exerts tumor-suppressive effects on cancers derived from epithelial cells. Although hypermethylation of CDH13 promoter has been reported in various cancers, its prognostic value for colorectal cancer (CRC) is still controversial. The methylation alterations of CDH13 within exon 1 have not yet been investigated. METHODS A total of 49 CRC patients were recruited for the prospective study. The methylation status of CpG sites was quantified by Bisulfite Amplicon Sequencing (BSAS) in malignant tissues and adjacent normal tissues. The primary endpoint of the study was overall survival (OS) after surgery. The relationship between methylation level with pathological stage and OS was also evaluated. RESULTS Compared with adjacent normal tissues, the overall average methylation level within exon 1 was significantly increased in tumor tissues (p < 0.001). The association study showed that the hypermethylation status of the CpG1 site was non-significantly associated with the presence of distant metastasis (p = 0.032). Moreover, the hypermethylation of two CpG sites, including CpG1 (p = 0.003) and CpG5 (p = 0.032), was associated with worse OS in CRC. Co-hypermethylation of CpG1 and CpG5 sites was significantly associated with a worse clinical outcome (HR: 4.43 [95% CI 1.27-15.46]; p = 0.019) in multivariate Cox regression analysis. CONCLUSION The methylation level of CDH13 exon 1 in CRC tissue was significantly higher than in adjacent normal tissues. Hypermethylation at the CpG1 site suggests a risk of distant metastasis in CRC. The hypermethylation of the CpG1 site and CpG5 site, including the co-hypermethylation of these two sites, may serve as a valuable prognostic biomarker.
Collapse
Affiliation(s)
- PengCheng Xiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - PengJu Li
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Xiaoqi Yuan
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Xiuhao Zhao
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Zitian Xiao
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Bingguan Chen
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Kenwen Liu
- Department of Gastrointestinal Surgery, Ji'an Central People's Hospital, Jian, 343000, Jiangxi, China
| | - Evelyne Bischof
- Department of Oncology and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Junyi Han
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China.
| |
Collapse
|
40
|
Wang X, Yang Z, Pu Z, Zheng Y, Chen H, Huang Y, Fan X, Yi P. Development and validation of a novel prognostic nomogram for hepatitis B virus-related acute-on-chronic liver failure patients receiving artificial liver therapy. Eur J Med Res 2024; 29:556. [PMID: 39568078 PMCID: PMC11580634 DOI: 10.1186/s40001-024-02141-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Hepatitis B virus-related acute-on-chronic liver failure (HBV-ACLF) is frequently accompanied by short-term morbidity and mortality. However, there have been no studies on the associations between baseline clinicopathologic characteristics at hospital admission and clinical prognosis after receiving artificial liver therapy. Therefore, the current study aimed to develop a prognostic nomogram for predicting the outcomes of patients with HBV-ACLF following artificial liver support. METHODS A retrospective study of 110 consecutive patients who were diagnosed with HBV-ACLF between January 2018 and August 2022 was conducted. First, univariate and multivariate logistic regression analyses were performed to determine the independent prognostic factors significantly associated with patient outcomes. Moreover, a predictive nomogram model underlying the prognostic factors was established and further evaluated. The area under the curve (AUC) was used to gauge the predictive accuracy. The calibration curve and decision curve analysis (DCA) were employed to assess the discriminability and clinical effectiveness, respectively. RESULTS In patients with HBV-ACLF, multivariate logistic analysis revealed that age ≥ 40 years (OR 6.76, p = 0.025), middle-stage liver failure (OR 49.96, p < 0.001), end-stage liver failure (OR 19.27, p = 0.002), hepatic encephalopathy (OR 7.06, p = 0.032), upper gastrointestinal hemorrhage (OR 47.24, p = 0.047), and artificial liver therapy consisting of plasma exchange (PE) + plasma exchange double plasma molecular adsorption system (DPMAS) (OR 0.26, p = 0.04) were identified as prognostic factors. Then, we established and evaluated a predictive nomogram with an AUC of 0.885, which showed better predictive accuracy than the model for end-stage liver disease (MELD) score (AUC of 0.634) and the Child-Pugh score (AUC of 0.611). Moreover, the calibration curve showed good agreement between the ideal and bias-corrected curves. Decision curve analysis confirmed the better clinical utility of this approach. CONCLUSIONS We developed and evaluated a unique nomogram that was more accurate than conventional prognostic models for predicting the clinical prognosis of HBV-ACLF patients receiving artificial liver therapy. As a result, the nomogram may be a helpful tool in clinical decision-making to predict the outcomes of patients with HBV-ACLF.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Infectious Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ziyue Yang
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Health Commission (NHC) Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhangya Pu
- Department of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang Province, China
| | - Yixiang Zheng
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Haiou Chen
- Department of Infectious Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Yan Huang
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xuegong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Panpan Yi
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
41
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Extracellular Vesicles in Viral Liver Diseases. Viruses 2024; 16:1785. [PMID: 39599900 PMCID: PMC11598962 DOI: 10.3390/v16111785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Extracellular vesicles (EVs) are bilayer vesicles released by cells in the microenvironment of the liver including parenchymal and non-parenchymal cells. They are the third important mechanism in the communications between cells, besides the secretion of cytokines and chemokines and the direct cell-to-cell contact. The aim of this review is to discuss the important role of EVs in viral liver disease, as there is increasing evidence that the transportation of viral proteins, all types of RNA, and viral particles including complete virions is implicated in the pathogenesis of both viral cirrhosis and viral-related hepatocellular carcinoma. The biogenesis of EVs is discussed and their role in the pathogenesis of viral liver diseases is presented. Their use as diagnostic and prognostic biomarkers is also analyzed. Most importantly, the significance of possible novel treatment strategies for liver fibrosis and hepatocellular carcinoma is presented, although available data are based on experimental evidence and clinical trials have not been reported.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
42
|
Antwi SO, Siaw ADJ, Armasu SM, Frank JA, Yan IK, Ahmed FY, Izquierdo-Sanchez L, Boix L, Rojasti A, Banales JM, Reig M, Stål P, Romero Gómez M, Wangensteen KJ, Singal AG, Roberts LR, Patel T. Genome-wide DNA methylation markers associated with metabolic liver cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.15.24317378. [PMID: 39606355 PMCID: PMC11601684 DOI: 10.1101/2024.11.15.24317378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background and Aims Metabolic liver disease is the fastest rising cause of hepatocellular carcinoma (HCC) worldwide, but the underlying molecular processes that drive HCC development in the setting of metabolic perturbations are unclear. We investigated the role of aberrant DNA methylation in metabolic HCC development in a multicenter international study. Methods We used a case-control design, frequency-matched on age, sex, and study site. Genome-wide profiling of peripheral blood leukocyte DNA was performed using the 850k EPIC array. Cell type proportions were estimated from the methylation data. The study samples were split 80% and 20% for training and validation. Differential methylation analysis was performed with adjustment for cell type, and we generated area under the receiver-operating curves (ROC-AUC). Results We enrolled 272 metabolic HCC patients and 316 control patients with metabolic liver disease from six sites. Fifty-five differentially methylated CpGs were identified; 33 hypermethylated and 22 hypomethylated in cases versus controls. The panel of 55 CpGs discriminated between cases and controls with AUC=0.79 (95%CI=0.71-0.87), sensitivity=0.77 (95%CI=0.66-0.89), and specificity=0.74 (95%CI=0.64-0.85). The 55-CpG classifier panel performed better than a base model that comprised age, sex, race, and diabetes mellitus (AUC=0.65, 95%CI=0.55-0.75, sensitivity=0.62 (95%CI=0.49-0.75) and specificity=0.64 (95%CI=0.52-0.75). A multifactorial model that combined the 55 CpGs with age, sex, race, and diabetes, yielded AUC=0.78 (95%CI=0.70-0.86), sensitivity=0.81 (95%CI=0.71-0.92), and specificity=0.67 (95%CI=0.55-0.78). Conclusions A panel of 55 blood leukocyte DNA methylation markers differentiates patients with metabolic HCC from control patients with benign metabolic liver disease, with a slightly higher sensitivity when combined with demographic and clinical information.
Collapse
Affiliation(s)
- Samuel O. Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Ampem Darko Jnr. Siaw
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Sebastian M. Armasu
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jacob A. Frank
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Irene K. Yan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Fowsiyo Y. Ahmed
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, San Sebastian, Spain
| | - Loreto Boix
- BCLC Group, Liver Unit, ICMDM, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain. Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Barcelona University, Barcelona, Spain
| | - Angela Rojasti
- SeLiver Group, UCM Digestive Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, San Sebastian, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Maria Reig
- BCLC Group, Liver Unit, ICMDM, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain. Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Barcelona University, Barcelona, Spain
| | - Per Stål
- Department of Upper GI Diseases, Karolinska University Hospital, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Romero Gómez
- SeLiver Group, UCM Digestive Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Kirk J. Wangensteen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amit G. Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tushar Patel
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
43
|
Małodobra-Mazur M, Ołdakowska M, Dobosz T. Exploring PPAR Gamma and PPAR Alpha's Regulation Role in Metabolism via Epigenetics Mechanism. Biomolecules 2024; 14:1445. [PMID: 39595621 PMCID: PMC11591816 DOI: 10.3390/biom14111445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to a family of nuclear receptors. To date, three types of PPARs, namely PPARα, PPARδ, and PPARγ, have been identified, demonstrating co-expression across numerous tissues. PPARγ is primarily distributed in adipose tissue, the colon, the immune system, and the retina, while PPARα is predominantly expressed in metabolic tissues such as brown adipose tissue, the liver, and the kidneys. Both PPARγ and PPARα play crucial roles in various cellular processes. Recent data suggest that the PPAR family, among other mechanisms, might also be regulated by epigenetic mechanisms. Our recent studies, alongside numerous others, have highlighted the pivotal roles of DNA methylation and histone modifications in the regulation of PPARγ and PPARα, implicating them in the deterioration of metabolic disorders via epigenetic mechanisms. This still not fully understood mechanism of regulation in the nuclear receptors family has been summarized and described in the present paper. The present review summarizes the available data on PPARγ and PPARα regulation via epigenetic mechanisms, elucidating the link between the development of metabolic disorders and the dysregulation of PPARγ and PPARα resulting from these mechanisms.
Collapse
Affiliation(s)
- Małgorzata Małodobra-Mazur
- Department of Forensic Science, Division of Molecular Techniques, Wroclaw Medical University, Sklodowskiej-Curie 52, 51-367 Wroclaw, Poland; (M.O.); (T.D.)
| | | | | |
Collapse
|
44
|
Ma Q, Li W, Wu W, Sun M. Exploring the active ingredients and mechanisms of Liujunzi decoction in treating hepatitis B: a study based on network pharmacology, molecular docking, and molecular dynamics simulations. Comput Methods Biomech Biomed Engin 2024:1-25. [PMID: 39534925 DOI: 10.1080/10255842.2024.2427117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Liujunzi decoction (LJZD) is commonly used to treat hepatitis B virus (HBV), though its active ingredients and mechanisms are not fully known. This study identified core targets and active components of LJZD for treating hepatitis B (HB) through network pharmacology, molecular docking, and molecular dynamics simulation. Screening from databases yielded 533 active components, 2619 targets for LJZD, and 2910 for HB, with 891 intersecting targets. STRING and CytoHubba analyses identified AR and VDR as core targets, with key pathways including PI3K-Akt and MAPK. The findings clarify LJZD's multicomponent, multitarget mechanisms, supporting its clinical application for HB treatment.
Collapse
Affiliation(s)
- Qing Ma
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Wenjun Li
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenying Wu
- Department of Pharmacy, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Mei Sun
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Dropmann A, Alex S, Schorn K, Tong C, Caccamo T, Godoy P, Ilkavets I, Liebe R, Gonzalez D, Hengstler JG, Piiper A, Quagliata L, Matter MS, Waidmann O, Finkelmeier F, Feng T, Weiss TS, Rahbari N, Birgin E, Rasbach E, Roessler S, Breuhahn K, Tóth M, Ebert MP, Dooley S, Hammad S, Meindl-Beinker NM. The TGF-β1 target WISP1 is highly expressed in liver cirrhosis and cirrhotic HCC microenvironment and involved in pro- and anti-tumorigenic effects. Biochem Biophys Res Commun 2024; 732:150409. [PMID: 39033550 DOI: 10.1016/j.bbrc.2024.150409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION WNT1-inducible signalling pathway protein 1 (WISP1) promotes progression of several tumor entities often correlating with worse prognosis. Here its expression regulation and role in the progression of chronic liver diseases (CLD) was investigated. METHODS WISP1 expression was analyzed in human HCC datasets, in biopsies and serum samples and an HCC patient tissue microarray (TMA) including correlation to clinicopathological parameters. Spatial distribution of WISP1 expression was determined using RNAscope analysis. Regulation of WISP1 expression was investigated in cytokine-stimulated primary mouse hepatocytes (PMH) by array analysis and qRT-PCR. Outcome of WISP1 stimulation was analyzed by IncuCyte S3-live cell imaging, qRT-PCR, and immunoblotting in murine AML12 cells. RESULTS In a TMA, high WISP1 expression was positively correlated with early HCC stages and male sex. Highest WISP1 expression levels were detected in patients with cirrhosis as compared to healthy individuals, patients with early fibrosis, and non-cirrhotic HCC in liver biopsies, expression datasets and serum samples. WISP1 transcripts were predominantly detected in hepatocytes of cirrhotic rather than tumorous liver tissue. High WISP1 expression was associated with better survival. In PMH, AML12 and HepaRG, WISP1 was identified as a specific TGF-β1 target gene. Accordingly, expression levels of both cytokines positively correlated in human HCC patient samples. WISP1-stimulation induced the expression of Bcl-xL, PCNA and p21 in AML12 cells. CONCLUSIONS WISP1 expression is induced by TGF-β1 in hepatocytes and is associated with cirrhotic liver disease. We propose a crucial role of WISP1 in balancing pro- and anti-tumorigenic effects during premalignant stages of CLD.
Collapse
Affiliation(s)
- Anne Dropmann
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Sophie Alex
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Katharina Schorn
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Chenhao Tong
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Tiziana Caccamo
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Patricio Godoy
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany; IfADo-Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Iryna Ilkavets
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Roman Liebe
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany; Clinic of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Daniela Gonzalez
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Jan G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Albrecht Piiper
- Department of Internal Medicine 1, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Luca Quagliata
- Institute of Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Matthias S Matter
- Institute of Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Oliver Waidmann
- Department of Internal Medicine 1, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Fabian Finkelmeier
- Department of Internal Medicine 1, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Teng Feng
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Thomas S Weiss
- Children's University Hospital (KUNO), Center for Liver Cell Research, University Hospital Regensburg, Josef-Engert-Straße 9, 93053, Regensburg, Germany
| | - Nuh Rahbari
- Department of Surgery and European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany; Department of General and Visceral Surgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Emrullah Birgin
- Department of Surgery and European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany; Department of General and Visceral Surgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Erik Rasbach
- Department of General and Visceral Surgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany; Department of Surgery, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Marcell Tóth
- Institute of Pathology, University Hospital Heidelberg, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Matthias P Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany; DKFZ-Hector Cancer Institute at the University Medical Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany; Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Seddik Hammad
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Nadja M Meindl-Beinker
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
46
|
Yang JR, Tian YX, Li JE, Zhang Y, Fan YC, Wang K. Mex3a promoter hypomethylation can be utilized to diagnose HBV-associated hepatocellular carcinoma: a randomized controlled trial. Front Pharmacol 2024; 15:1325869. [PMID: 39564121 PMCID: PMC11574524 DOI: 10.3389/fphar.2024.1325869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma remains a health challenge for humanity. Therefore, there is an urgent need to develop novel biomarkers with high efficiency yet fast ability to meet the requirements of hepatocellular carcinoma treatment. METHODS A total of 229 patients with HBV-associated hepatocellular carcinoma (HCC), 298 patients with chronic hepatitis B (CHB), and 96 healthy controls were retrospectively analyzed. Methylation levels of the Mex3a promoter in peripheral blood mononuclear cells (PBMCs) were measured using MethyLight to obtain clinical and laboratory parameters. RESULTS The Mex3a promoter methylation level in HCC patients (median: 0.289% and interquartile range: 0.126%-0.590%) was significantly lower than that in CHB patients (median: 0.999%, interquartile range: 0.417%-1.268%, and p < 0.001) and healthy people (median: 2.172%, interquartile range: 1.225%-3.098%, and p < 0.001). The Mex3a mRNA levels in HCC patients (median: 12.198 and interquartile range: 3.112-18.996) were significantly higher than those in CHB patients (median: 1.623 and interquartile range: 0.066-6.000, and p < 0.001) and healthy controls (median: 0.329, interquartile range: 0.031-1.547, and p < 0.001). MethyLight data were expressed as a percentage of the methylated reference (PMR) value. The Mex3a PMR value was negatively correlated with the mRNA expression level (Spearman's R = -0.829 and p < 0.001). The Mex3a PMR value of HCC patients was significantly correlated with age (Spearman's R = 0.113 and p = 0.044), and the mRNA level was significantly correlated with ALT (Spearman's R = 0.132 and p = 0.046). The Mex3a promoter methylation levels and mRNA levels were also independent factors in the development of liver cancer. The Mex3a promoter methylation and mRNA levels were better at distinguishing HCC from CHB than AFP [area under the receiver operating characteristic curve (AUC) for predicting HCC vs. CHB: 0.915 vs. 0.715: p < 0.001]. The combined use of AFP and Mex3a methylation levels and mRNA levels further improved the area under the receiver operating characteristic curve. CONCLUSION The presence of Mex3a promoter hypomethylation in hepatocellular carcinoma can be used as a non-invasive biomarker for the early detection of liver cancer.
Collapse
Affiliation(s)
- Jie-Ru Yang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Xin Tian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Jin-E. Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Ying Zhang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| |
Collapse
|
47
|
Cheng Y, Song Z, Liu Y, Xu X, Zhang D, Zou Y, Liu L, Zeng Y, Li W, Bai D, Dai D. Common molecular basis for MASH and hepatitis C revealed via systems biology approach. Front Oncol 2024; 14:1442221. [PMID: 39605886 PMCID: PMC11599856 DOI: 10.3389/fonc.2024.1442221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Background Metabolic dysfunction-associated steatohepatitis (MASH) is characterized by liver inflammation and damage caused by a buildup of fat in the liver. Hepatitis C, caused by hepatitis C virus (HCV), is a disease that can lead to liver cirrhosis, liver cancer, and liver failure. MASH and hepatitis C are the common causes of liver cirrhosis and hepatocellular carcinoma. Several studies have shown that hepatic steatosis is also a common histological feature of liver in HCV infected patients. However, the common molecular basis for MASH and hepatitis C remains poorly understood. Methods Firstly, differentially expressed genes (DEGs) for MASH and hepatitis C were extracted from the GSE89632, GSE164760 and GSE14323 datasets. Subsequently, the common DEGs shared among these datasets were determined using the Venn diagram. Next, a protein-protein interaction (PPI) network was constructed based on the common DEGs and the hub genes were extracted. Then, gene ontology (GO) and pathway analysis of the common DEGs were performed. Furthermore, transcription factors (TFs) and miRNAs regulatory networks were constructed, and drug candidates were identified. After the MASH and hepatitis C cell model was treated with predicted drug, the expression levels of the signature genes were measured by qRT-PCR and ELISA. Results 866 common DEGs were identified in MASH and hepatitis C. The GO analysis showed that the most significantly enriched biological process of the DEGs was the positive regulation of cytokine production. 10 hub genes, including STAT1, CCL2, ITGAM, PTPRC, CXCL9, IL15, SELL, VCAM1, TLR4 and CCL5, were selected from the PPI network. By constructing the TF-gene and miRNA-gene network, most prominent TFs and miRNAs were screened out. Potential drugs screening shows that Budesonide and Dinoprostone may benefit patients, and cellular experiments showed that Budesonide effectively inhibited the expression of genes related to glycolipid metabolism, fibrosis, and inflammatory factors. Conclusion We extracted 10 hub genes between MASH and hepatitis C, and performed a series of analyses on the genes. Molecular docking and in vitro studies have revealed that Budesonide can effectively suppress the progression of MASH and hepatitis C. This study can provide novel insights into the potential drug targets and biomarkers for MASH and hepatitis C.
Collapse
Affiliation(s)
- Yongwei Cheng
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Zihao Song
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Ye Liu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Xichao Xu
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Dali Zhang
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yigui Zou
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Liang Liu
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yinzhen Zeng
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Wenwen Li
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Daming Bai
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| | - Dongling Dai
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Endoscopy Center and Gastroenterology Department, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
48
|
Su L, Bu J, Yu J, Jin M, Meng G, Zhu X. Comprehensive review and updated analysis of DNA methylation in hepatocellular carcinoma: From basic research to clinical application. Clin Transl Med 2024; 14:e70066. [PMID: 39462685 PMCID: PMC11513202 DOI: 10.1002/ctm2.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary malignant tumour, ranking second in global mortality rates and posing significant health threats. Epigenetic alterations, particularly DNA methylation, have emerged as pivotal factors associated with HCC diagnosis, therapy, prognosis and malignant progression. However, a comprehensive analysis of the DNA methylation mechanism driving HCC progression and its potential as a therapeutic biomarker remains lacking. This review attempts to comprehensively summarise various aspects of DNA methylation, such as its mechanism, detection methods and biomarkers aiding in HCC diagnosis, treatment and prognostic assessment of HCC. It also explores the role of DNA methylation in regulating HCC's malignant progression and sorafenib resistance, alongside elaborating the therapeutic effects of DNA methyltransferase inhibitors on HCC. A detailed examination of these aspects underscores the significant research on DNA methylation in tumour cells to elucidate malignant progression mechanisms, identify diagnostic markers and develop new tumour-specific inhibitors for HCC. KEY POINTS: A comprehensive summary of various aspects of DNA methylation, such as its mechanism, detection methods and biomarkers aiding in diagnosis and treatment. The role of DNA methylation in regulating hepatocellular carcinoma's (HCC) malignant progression and sorafenib resistance, alongside elaborating therapeutic effects of DNA methyltransferase inhibitors. Deep research on DNA methylation is critical for discovering novel tumour-specific inhibitors for HCC.
Collapse
Affiliation(s)
- Lin Su
- Department of Pain ManagementShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiawen Bu
- Department of Colorectal SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiahui Yu
- Department of UltrasoundShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Mila Jin
- Department of Operation RoomThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Guanliang Meng
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xudong Zhu
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Department of General SurgeryCancer Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
49
|
Bai T, Mai RY, Tang ZH, Wang XB, Chen J, Ye JZ, Wei M, Zhang B, Li K, Gu ZM, Wu FX, Li LQ. Ubiquitination of P53 Regulated by Ubiquitin-Specific Protease 14 Delays the Invasion of Hepatitis B Virus and the Development of Hepatitis. Viral Immunol 2024; 37:432-439. [PMID: 39601371 DOI: 10.1089/vim.2024.0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
This study aims to explore the mechanism underlying the role of ubiquitin-specific protease 14 (USP14) in regulating P53 expression and influencing the development of hepatitis B. The animal and cell models of hepatitis B were constructed. The mRNA and protein expression of USP14, mouse double minute 2 (MDM2), and P53 were detected by western blot and qPCR. The USP14 overexpression vector was constructed. The pathological changes of liver tissue were detected by HE and Masson staining. Protein immunoprecipitation was used to detect the interaction between MDM2 and P53, as well as between MDM2 and USP14. The ubiquitination levels of P53 after USP14 overexpression were detected. qPCR and western blot were used to detect the expression of MDM2, Bcl-2, P53, Bax, and Caspase-1 in vivo and in vitro. Compared with the control group, the model group showed increased cell proliferation, increased expression of MDM2 and Bcl-2 in cells and liver tissue, and decreased expression of P53, Bax, and Caspase-1. Compared with the model group, overexpression of USP14 resulted in a decrease in MDM2 expression and an increase in P53 expression. After transfection with the USP14 overexpression plasmid, cell proliferation was inhibited, and the expression of MDM2 and Bcl-2 was decreased in cells and liver tissue, while the expression of P53, Bax, and Caspase-1 was increased. In the model of hepatitis B, USP14 upregulation downregulated MDM2 and promoted P53 deubiquitination to delay the invasion of hepatitis B virus and the development of hepatitis.
Collapse
Affiliation(s)
- Tao Bai
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Rong-Yun Mai
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhi-Hong Tang
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiao-Bo Wang
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jie Chen
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jia-Zhou Ye
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Meng Wei
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bin Zhang
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Kai Li
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhao-Min Gu
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fei-Xiang Wu
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Le-Qun Li
- Hepatobiliary Pancreatic Splenic Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
50
|
Hetta HF, Hamed HM, Mekky MA, Abdel-Malek MO, Hassan WA. Circulating microRNA-21, microRNA-122, and microRNA-222 as diagnostic biomarkers for hepatitis c virus-related hepatocellular carcinoma. EGYPTIAN LIVER JOURNAL 2024; 14:78. [DOI: 10.1186/s43066-024-00385-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/27/2024] [Indexed: 01/03/2025] Open
Abstract
Abstract
Background and aim
MicroRNAs (miRs) are now a well-known subject in various tumor genesis and are studied as early diagnostic biomarker. Many arrays of miRs were incorporated in the pathogenesis of HCV-related hepatocellular carcinomas (HCV-HCC). In this respect, we aimed to evaluate the diagnostic role of circulating miR-21, miR-122, and miR-222 in Egyptian patients with HCV-HCC.
Patient and methods
Between June 2018 and April 2019, a cross-sectional comparative study was designed to evaluate the circulating miR-21, miR-122, and miR-222 by quantitative Real-Time PCR. For analytical purposes, patients were categorized into three groups: chronic HCV group (CHC-group, n = 22), HCV-related liver cirrhosis (LC-group, n = 22), and HCV-related hepatocellular carcinoma (HCV-HCC-group, n = 54).
Results
Serum levels of miR-21 and miR-222 increased with the progressive course from CHC to LC and HCC; p < .001. Serum levels of miR-122 in HCC patients were significantly lower than non-HCC patients (CHC and LC patients, n = 44); p < .001. However, the differences in levels of serum miR-122 between CHC and LC were not statistically significant; P = 0.8.
ROC curve analysis showed that the sensitivity and specificity of miR-21 were 61.1% and 95.5%, miR-222 were 71.7% and 93.2%, and miR-122 were 98.2% and 100%. The positive predictive value for miRNA-21, miRNA-122, and miRNA-222 were 13.4%, 93.3%, and 10.5% respectively. The Negative predictive value for miRNA-21, miRNA-122, and miRNA-222 were 94.3%, 97.8%, and 92.7% respectively.
Conclusion
MiR-21 and miR-222 could be potential markers for advanced liver damage, while miR-122 had the best diagnostic accuracy and could be a promising marker for detection of HCC.
Collapse
|